US20240050569A1 - Mesothelin binding molecules and uses thereof - Google Patents

Mesothelin binding molecules and uses thereof Download PDF

Info

Publication number
US20240050569A1
US20240050569A1 US18/268,215 US202118268215A US2024050569A1 US 20240050569 A1 US20240050569 A1 US 20240050569A1 US 202118268215 A US202118268215 A US 202118268215A US 2024050569 A1 US2024050569 A1 US 2024050569A1
Authority
US
United States
Prior art keywords
sequence
cdr
seq
cancer
car
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US18/268,215
Inventor
Farzad Haerizadeh
Masood Tayebi
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Bioardis LLC
Original Assignee
Bioardis LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bioardis LLC filed Critical Bioardis LLC
Priority to US18/268,215 priority Critical patent/US20240050569A1/en
Assigned to BIOARDIS, LLC reassignment BIOARDIS, LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HAERIZADEH, FARZAD, TAYEBI, MASOOD
Publication of US20240050569A1 publication Critical patent/US20240050569A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464466Adhesion molecules, e.g. NRCAM, EpCAM or cadherins
    • A61K39/464468Mesothelin [MSLN]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2302Interleukin-2 (IL-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2307Interleukin-7 (IL-7)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes

Definitions

  • MSLN mesothelin
  • CAR chimeric antigen receptor
  • Chimeric antigen receptor (CAR) T cells and other adoptive cell therapies have been shown to be effective in the treatment of cancer.
  • the CAR which is made up of an extracellular antigen binding domain, a transmembrane domain, and an intracellular signaling domain, enables directed killing of cancer cells based on cell surface antigen expression while minimally affecting normal cells that are not expressing the targeted antigen.
  • the extracellular antigen binding domain is often made up of an antibody or a binding fragment or derivative thereof, such as a single chain variable fragment (scFv) or single domain antibody (sdAb).
  • scFv single chain variable fragment
  • sdAb single domain antibody
  • binding polypeptides that are able to bind to mesothelin (MSLN). These binding polypeptides may be incorporated in a chimeric antigen receptor (CAR), which can be expressed by a cell. In some embodiments, the binding polypeptides are single domain antibodies (sdAbs).
  • MSLN binding polypeptides comprising an immunoglobulin heavy chain variable domain comprising a CDR-H1, CDR-H2, and CDR-H3.
  • the CDR-H1 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 1-41.
  • CDR-H2 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 42-82.
  • the CDR-H3 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 83-123.
  • the CDR-H1 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 1-41
  • the CDR-H2 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 42-82
  • the CDR-H3 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 83-123.
  • the immunoglobulin heavy chain variable domain comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 124-164.
  • nucleic acids that encode for any one of the MSLN binding polypeptides disclosed herein.
  • the methods comprise administering a chimeric antigen receptor cell to the subject.
  • the chimeric antigen receptor cell is any one of the chimeric antigen receptor cells disclosed herein.
  • the chimeric antigen receptor cell comprises any one or more of the MSLN binding polypeptides disclosed herein.
  • a mesothelin (MSLN) binding polypeptide comprising an immunoglobulin heavy chain variable domain comprising a CDR-H1, CDR-H2, and CDR-H3, wherein:
  • sdAb single domain antibody
  • a chimeric antigen receptor (CAR) comprising the MSLN binding polypeptide of any one of alternatives 1-5.
  • a chimeric antigen receptor (CAR) cell comprising the CAR of alternative 6.
  • cancer is breast cancer, colorectal cancer, kidney cancer, liver cancer, lung cancer, brain cancer, pancreatic cancer, bladder cancer, testicular cancer, prostate cancer, gastric cancer, ovarian cancer, head and neck cancer, gallbladder cancer, a hematologic malignancy, or any combination thereof.
  • FIG. 1 depicts an exemplary alignment for the heavy chain variable domain CDRs disclosed herein.
  • FIG. 2 A depicts a bar chart which shows the results of a cytotoxicity assay of exemplary anti-MSLN CAR T-cell lines against BxPC-3 pancreatic cancer cells.
  • FIG. 2 B depicts a bar chart which shows the results of a cytotoxicity assay of exemplary anti-MSLN CAR T-cell lines against Capan-1 pancreatic cancer cells.
  • FIG. 2 C depicts a bar chart which shows the results of a cytotoxicity assay of exemplary anti-MSLN CAR T-cell lines against NCI-N87 gastric cancer cells, where the T-cells have been obtained from a donor denoted Z0016.
  • FIG. 2 D depicts a bar chart which shows the results of a cytotoxicity assay of exemplary anti-MSLN CAR T-cell lines against NCI-N87 gastric cancer cells, where the T-cell have been obtained from a donor denoted Y1287.
  • FIG. 3 depicts three line graphs which show the anti-tumor efficacy of an exemplary anti-MSLN CAR T-cell line B4T2-002 ('002) against HCC1806 breast cancer, H2110 non-small cell lung cancer, and NCI-N87 gastric cancer xenograph models.
  • binding polypeptides that are incorporated into a chimeric antigen receptor cell.
  • the chimeric antigen receptor cell is a chimeric antigen receptor T cell (CAR-T cell).
  • CAR-Ts may be constructed through processes conventionally known in the art.
  • the binding polypeptides provide specificity towards their respective tumor-associated antigens, enabling targeting of cancers expressing said tumor-associated antigens by the CAR-T cell.
  • the binding polypeptides are single domain antibodies (sdAbs) disposed on the surface of the chimeric antigen receptor cells (e.g. CAR-T cell).
  • the sdAbs may be specific for, or have binding affinity towards, a tumor-associated antigen.
  • the tumor-associated antigen is mesothelin (MSLN).
  • the cancer may be breast cancer, colorectal cancer, kidney cancer, liver cancer, lung cancer, brain cancer, pancreatic cancer, bladder cancer, testicular cancer, prostate cancer, gastric cancer, ovarian cancer, head and neck cancer, gallbladder cancer, a hematologic malignancy, or any combination thereof.
  • the hematologic malignancy may comprise leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, hairy cell leukemia, lymphoma, Hodgkin's disease, Non-Hodgkin lymphoma, or multiple myeloma.
  • the CAR-T cell may be derived from the subject for an autologous treatment. Alternatively, the CAR-T cell may be derived from the same species as the subject for an allogeneic treatment.
  • an element means one element or more than one element.
  • the terms “individual(s)”, “subject(s)” and “patient(s)” mean any mammal.
  • the mammal is a human.
  • the mammal is a non-human. None of the terms require or are limited to situations characterized by the supervision (e.g. constant or intermittent) of a health care worker (e.g. a doctor, a registered nurse, a nurse practitioner, a physician's assistant, an orderly or a hospice worker).
  • a health care worker e.g. a doctor, a registered nurse, a nurse practitioner, a physician's assistant, an orderly or a hospice worker.
  • administering includes enteral, oral, intranasal, parenteral, intravenous, intraperitoneal, intramuscular, intra-arteriole, intraventricular, intradermal, intralesional, intracranial, intrathecal, or subcutaneous administration, or the implantation of a slow-release device, e.g., a mini-osmotic pump, to a subject.
  • a slow-release device e.g., a mini-osmotic pump
  • nucleic acid or “nucleic acid molecule” as used herein refers to polynucleotides, such as deoxyribonucleic acid (DNA) or ribonucleic acid (RNA), oligonucleotides, fragments generated by the polymerase chain reaction (PCR), and fragments generated by any of ligation, scission, endonuclease action, and exonuclease action.
  • Nucleic acid molecules can be composed of monomers that are naturally-occurring nucleotides (such as DNA and RNA), or analogs of naturally-occurring nucleotides (e.g., enantiomeric forms of naturally-occurring nucleotides), or a combination of both.
  • Nucleic acid monomers can be linked by phosphodiester bonds or analogs of such linkages.
  • a nucleic acid or nucleic acids can be contained in a nucleic acid vector or nucleic acid construct (e.g. plasmid, virus, bacteriophage, cosmid, fosmid, phagemid, bacterial artificial chromosome (BAC), yeast artificial chromosome (YAC), or human artificial chromosome (HAC)) that can be used for amplification and/or expression of the nucleic acid or nucleic acids in various biological systems.
  • a nucleic acid vector or nucleic acid construct e.g. plasmid, virus, bacteriophage, cosmid, fosmid, phagemid, bacterial artificial chromosome (BAC), yeast artificial chromosome (YAC), or human artificial chromosome (HAC)
  • the vector or construct will also contain elements including but not limited to promoters, enhancers, terminators, inducers, ribosome binding sites, translation initiation sites, start codons, stop codons, polyadenylation signals, origins of replication, cloning sites, multiple cloning sites, restriction enzyme sites, epitopes, reporter genes, selection markers, antibiotic selection markers, targeting sequences, peptide purification tags, or accessory genes, or any combination thereof.
  • elements including but not limited to promoters, enhancers, terminators, inducers, ribosome binding sites, translation initiation sites, start codons, stop codons, polyadenylation signals, origins of replication, cloning sites, multiple cloning sites, restriction enzyme sites, epitopes, reporter genes, selection markers, antibiotic selection markers, targeting sequences, peptide purification tags, or accessory genes, or any combination thereof.
  • a nucleic acid or nucleic acid molecule can comprise one or more sequences encoding different peptides, polypeptides, or proteins. These one or more sequences can be joined in the same nucleic acid or nucleic acid molecule adjacently, or with extra nucleic acids in between, e.g. linkers, repeats or restriction enzyme sites, or any other sequence that is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, or 300 bases long, or any length in a range defined by any two of the aforementioned lengths.
  • downstream on a nucleic acid as used herein refers to a sequence being after the 3′-end of a previous sequence, on the strand containing the encoding sequence (sense strand) if the nucleic acid is double stranded.
  • upstream on a nucleic acid as used herein refers to a sequence being before the 5′-end of a subsequent sequence, on the strand containing the encoding sequence (sense strand) if the nucleic acid is double stranded.
  • grouped on a nucleic acid as used herein refers to two or more sequences that occur in proximity either directly or with extra nucleic acids in between, e.g.
  • linkers repeats, or restriction enzyme sites, or any other sequence that is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, or 300 bases long, or any length in a range defined by any two of the aforementioned lengths, but generally not with a sequence in between that encodes for a functioning or catalytic polypeptide, protein, or protein domain.
  • codon optimized refers to the substitution of codons of the nucleic acid to enhance or maximize translation in a host of a particular species without changing the polypeptide sequence based on species-specific codon usage biases and relative availability of each aminoacyl-tRNA in the target cell cytoplasm. Codon optimization and techniques to perform such optimization is known in the art. Those skilled in the art will appreciate that gene expression levels are dependent on many factors, such as promoter sequences and regulatory elements. In this aspect, many synthetic genes can be designed to increase their protein expression level.
  • peptide refers to macromolecules comprised of amino acids linked by peptide bonds.
  • the numerous functions of peptides, polypeptides, and proteins are known in the art, and include but are not limited to enzymes, structure, transport, defense, hormones, or signaling. Peptides, polypeptides, and proteins are often, but not always, produced biologically by a ribosomal complex using a nucleic acid template, although chemical syntheses are also available.
  • nucleic acid template By manipulating the nucleic acid template, peptide, polypeptide, and protein mutations such as substitutions, deletions, truncations, additions, duplications, or fusions of more than one peptide, polypeptide, or protein can be performed. These fusions of more than one peptide, polypeptide, or protein can be joined in the same molecule adjacently, or with extra amino acids in between, e.g.
  • linkers repeats, epitopes, or tags, or any other sequence that is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, or 300 bases long, or any length in a range defined by any two of the aforementioned lengths.
  • downstream on a polypeptide as used herein refers to a sequence being after the C-terminus of a previous sequence.
  • upstream on a polypeptide as used herein refers to a sequence being before the N-terminus of a subsequent sequence.
  • nucleic acid or peptide sequences presented herein and used in the examples are functional in various biological systems including but not limited to humans, mice, rats, monkeys, primates, cats, dogs, rabbits, E. coli , yeast, and mammalian cells.
  • nucleic acid or peptide sequences sharing at least or lower than 0%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity, or any percentage within a range defined by any two of the aforementioned percentages of identity to the nucleic acid or peptide sequences presented herein and used in the examples can also be used with little or no effect on the function of the sequences in biological systems.
  • identity refers to a nucleic acid or peptide sequence having the same overall order of nucleotide or amino acids, respectively, as a template nucleic acid or peptide sequence with specific changes such as substitutions, deletions, repetitions, or insertions within the sequence.
  • two nucleic acid sequences sharing as low as 0%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity can encode for the same polypeptide by comprising different codons that encode for the same amino acid during translation.
  • sequences having a % homology to any of the sequences disclosed herein are envisioned and may be used.
  • the term “% homology” refers to the degree of conservation between two sequences when considering their three-dimensional structure. For example, homology between two protein sequences may be dependent on structural motifs, such as beta strands, alpha helices, and other folds, as well as their distribution throughout the sequence. Homology may be determined through structural determination, either empirically or in silico.
  • any sequence having at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence homology to any of the sequences disclosed herein may be used.
  • any sequence having at least 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 substitutions, deletions, or additions relative to any of the sequences disclosed herein, which may or may not affect the overall % homology, may be used.
  • sequences having a certain % similarity to any of the sequence disclosed herein are envisioned and may be used.
  • these sequences may include peptide sequences, nucleic acid sequences, CDR sequences, variable region sequences, or heavy or light chain sequences.
  • similarity refers to the comparison of amino acids based on their properties, including but not limited to size, polarity, charge, pK, aromaticity, hydrogen bonding properties, or presence of functional groups (e.g. hydroxyl, thiol, amine, carboxyl, and the like).
  • % similarity refers to the percentage of units (i.e.
  • amino acids that are the same between two or more sequences relative to the length of the sequence.
  • the % similarity will be respective that length.
  • deletions and/or insertions may be introduced to obtain the best alignment.
  • the similarity of two amino acids may dictate whether a certain substitution is conservative or non-conservative. Methods of determining the conservativeness of an amino acid substitution are generally known in the art and may involve substitution matrices.
  • substitution matrices include BLOSUM45, BLOSUM62, BLOSUM80, PAM100, PAM120, PAM160, PAM200, PAM250, but other substitution matrices or approaches may be used as considered appropriate by the skilled person.
  • a certain substitution matrix may be preferential over the others when considering aspects such as stringency, conservation and/or divergence of related sequences (e.g. within the same species or broader), and length of the sequences in question.
  • a peptide sequence having a certain % similarity to another sequence will have up to that % of amino acids that are either identical or an acceptable substitution as governed by the method of similarity determination used.
  • a sequence having at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence similarity to any of the sequences disclosed herein may be used.
  • any sequence having at least 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 similar substitutions relative to any of the sequences disclosed herein may be used.
  • these similar substitutions may apply to antigen-binding regions (i.e. CDRs) or regions that do not bind to antigens or are only secondary to antigen binding (i.e. framework regions).
  • consensus sequence refers to the generalized sequence representing all of the different combinations of permissible amino acids at each location of a group of sequences.
  • a consensus sequence may provide insight into the conserved regions of related sequences where the unit (e.g. amino acid or nucleotide) is the same in most or all of the sequences, and regions that exhibit divergence between sequences.
  • the consensus sequence of a CDR may indicate amino acids that are important or dispensable for antigen binding. It is envisioned that consensus sequences may be prepared with any of the sequences provided herein, and the resultant various sequences derived from the consensus sequence can be validated to have similar effects as the template sequences.
  • antibody denotes the meaning ascribed to it by one of skill in the art, and further it is intended to include any polypeptide chain-containing molecular structure with a specific shape that fits to and recognizes an epitope, where one or more non-covalent binding interactions stabilize the complex between the molecular structure and the epitope.
  • antibody library refers to a collection of antibodies and/or antibody fragments displayed for screening and/or combination into full antibodies.
  • the antibodies and/or antibody fragments may be displayed on a ribosome; on a phage; or on a cell surface, in particular a yeast cell surface.
  • the term “compete,” as used herein with regard to an antibody or binding polypeptide, means that a first antibody or binding polypeptide, or an antigen-binding portion thereof, binds to an epitope in a manner sufficiently similar to the binding of a second antibody or binding polypeptide, or an antigen-binding portion thereof, such that the result of binding of the first antibody or binding polypeptide with its cognate epitope is detectably decreased in the presence of the second antibody or binding polypeptide compared to the binding of the first antibody or binding polypeptide in the absence of the second antibody or binding polypeptide.
  • the alternative, where the binding of the second antibody or binding polypeptide to its epitope is also detectably decreased in the presence of the first antibody or binding polypeptide can, but need not be the case.
  • An antibody or binding polypeptide that “preferentially binds” or “specifically binds” (used interchangeably herein) to an epitope is a term well understood in the art, and methods to determine such specific or preferential binding are also well known in the art.
  • a molecule is said to exhibit “specific binding” or “preferential binding” if it reacts or associates more frequently, and/or more rapidly, and/or with greater duration and/or with greater affinity with a particular cell or substance than it does with alternative cells or substances.
  • An antibody or binding polypeptide “specifically binds” or “preferentially binds” to a target if it binds with greater affinity, and/or avidity, and/or more readily, and/or with greater duration than it binds to other substances.
  • non-human antibodies are hybrid immunoglobulins, immunoglobulin chains or fragments thereof which contain minimal sequence derived from non-human immunoglobulin.
  • single domain binding polypeptide or “single domain antibody” (sdAb) as used herein refers to a single peptide strand (e.g. not bound to another peptide strand with disulfide bonds) comprising an intact immunoglobulin domain or other protein fold which can recognize antigens.
  • Single domain binding polypeptides or sdAbs may be derived from typical heavy or light immunoglobulin chains, such as from human, or from alternative sources such as dromedaries (e.g. V H H) and cartilaginous fish (e.g. V NAR ).
  • the single domain binding polypeptide or sdAb comprises one, two, or three complementarity determining regions (CDRs).
  • the single domain binding polypeptide or sdAb comprises one, two, or three of a CDR1, CDR2, and CDR3.
  • single-chain variable fragment as used herein is a fusion protein comprising the variable regions of the heavy (VH) and light chains (VL) of an immunoglobulin, in which the VH and VL are covalently linked to form a VH:VL heterodimer.
  • the VH and VL are either joined directly or joined by a peptide-encoding linker, which connects the N-terminus of the VH with the C-terminus of the VL, or the C-terminus of the VH with the N-terminus of the VL.
  • the linker is usually rich in glycine for flexibility, as well as serine or threonine for solubility.
  • Single chain Fv polypeptide antibodies can be expressed from a nucleic acid including VH- and VL-encoding sequences.
  • the VH and VL of the scFv each comprises one, two, or three CDRs.
  • the VH and VL of the scFv each comprises one, two, or three of a CDR1, CDR2, and CDR3.
  • definitive delineation of a CDR and identification of residues comprising the binding site of an antibody or binding polypeptide is accomplished by solving the structure of the antibody or binding polypeptide and/or solving the structure of the antibody-ligand complex. In certain embodiments, that can be accomplished by any of a variety of techniques known to those skilled in the art, such as X-ray crystallography.
  • various methods of analysis can be employed to identify or approximate the CDR regions. In certain embodiments, various methods of analysis can be employed to identify or approximate the CDR regions. Examples of such methods include, but are not limited to, the Kabat definition, the Chothia definition, the IMGT approach (Lefranc et al., 2003) Dev Comp Immunol. 27:55-77), computational programs such as Paratome (Kunik et al., 2012, Nucl Acids Res. W521-4), the AbM definition, and the conformational definition.
  • the Kabat definition is a standard for numbering the residues in an antibody and is typically used to identify CDR regions. See, e.g., Johnson & Wu, 2000, Nucleic Acids Res., 28:214-8.
  • the Chothia definition is similar to the Kabat definition, but the Chothia definition takes into account positions of certain structural loop regions. See, e.g., Chothia et al., 1986, J. Mol. Biol., 196:901-17; Chothia et al., 1989, Nature, 342:877-83.
  • the AbM definition uses an integrated suite of computer programs produced by Oxford Molecular Group that model antibody structure.
  • the AbM definition models the tertiary structure of an antibody from primary sequence using a combination of knowledge databases and ab initio methods, such as those described by Samudrala et al., 1999, “Ab Initio Protein Structure Prediction Using a Combined Hierarchical Approach,” in PROTEINS, Structure, Function and Genetics Suppl., 3:194-198.
  • the contact definition is based on an analysis of the available complex crystal structures.
  • CDRs In another approach, referred to herein as the “conformational definition” of CDRs, the positions of the CDRs may be identified as the residues that make enthalpic contributions to antigen binding. See, e.g., Makabe et al., 2008, Journal of Biological Chemistry, 283:1156-1166. Still other CDR boundary definitions may not strictly follow one of the above approaches, but will nonetheless overlap with at least a portion of the Kabat CDRs, although they may be shortened or lengthened in light of prediction or experimental findings that particular residues or groups of residues do not significantly impact antigen binding.
  • a CDR may refer to CDRs defined by any approach known in the art, including combinations of approaches.
  • the methods used herein may utilize CDRs defined according to any of these approaches.
  • the CDRs may be defined in accordance with any of Kabat, Chothia, extended, IMGT, Paratome, AbM, and/or conformational definitions, or a combination of any of the foregoing.
  • CAR chimeric antigen receptor
  • An exemplary immune cell in which CARs can be used are T cells, but it is envisioned that CARs can be engineered into any amenable cytotoxic immune cell, including but not limited to T cells, Natural Killer (NK) cells, Natural Killer T (NKT) cells, dendritic cells, or macrophages.
  • NK Natural Killer
  • NKT Natural Killer T
  • dendritic cells dendritic cells
  • macrophages any disclosure pertaining to CAR T cells can also be applied to other immune cells comprising CARs.
  • CARs comprise an extracellular antigen-recognizing domain (e.g. tumor receptor ligand, or antibody), hinge, transmembrane, and intracellular signaling domain (endodomain). Different combinations of these CAR components may result in different specificities and efficacy against certain cancer antigens.
  • treating means an approach for obtaining beneficial or desired results in a subject's condition, including clinical results.
  • beneficial or desired clinical results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of the extent of a disease, stabilizing (i.e., not worsening) the state of disease, prevention of a disease's transmission or spread, delaying or slowing of disease progression, amelioration or palliation of the disease state, diminishment of the reoccurrence of disease, and remission, whether partial or total and whether detectable or undetectable.
  • “Treating” and “treatment” as used herein also include prophylactic treatment.
  • Treatment methods comprise administering to a subject a therapeutically effective amount of an active agent.
  • the administering step may consist of a single administration or may comprise a series of administrations.
  • the compositions are administered to the subject in an amount and for a duration sufficient to treat the subject.
  • the length of the treatment period depends on a variety of factors, such as the severity of the condition, the age and genetic profile of the subject, the concentration of active agent, the activity of the compositions used in the treatment, or a combination thereof.
  • the effective dosage of an agent used for the treatment or prophylaxis may increase or decrease over the course of a particular treatment or prophylaxis regime. Changes in dosage may result and become apparent by standard diagnostic assays known in the art. In some instances, chronic administration may be required.
  • an effective amount refers to that amount of a recited composition or compound that results in an observable designated effect.
  • Actual dosage levels of active ingredients in an active composition of the presently disclosed subject matter can be varied so as to administer an amount of the active composition or compound that is effective to achieve the designated response for a particular subject and/or application.
  • the selected dosage level can vary based upon a variety of factors including, but not limited to, the activity of the composition, formulation, route of administration, combination with other drugs or treatments, severity of the condition being treated, and the physical condition and prior medical history of the subject being treated.
  • a minimal dose is administered, and dose is escalated in the absence of dose-limiting toxicity to a minimally effective amount. Determination and adjustment of an effective dose, as well as evaluation of when and how to make such adjustments, are contemplated herein.
  • administering includes oral administration, topical contact, administration as a suppository, parenteral, intravenous, intraperitoneal, intramuscular, intralesional, intrathecal, intranasal, subdermal, or subcutaneous administration, or the implantation of a slow-release device, e.g., a mini-osmotic pump, to a subject.
  • Administration is by any route, including parenteral and transmucosal (e.g., buccal, sublingual, palatal, gingival, nasal, vaginal, rectal, or transdermal).
  • Parenteral administration includes, e.g., intravenous, intramuscular, intra-arteriole, intradermal, subcutaneous, intraperitoneal, intraventricular, and intracranial.
  • Other modes of delivery include, but are not limited to, the use of liposomal formulations, intravenous infusion, transdermal patches, etc.
  • co-administer it is meant that a first compound described herein is administered at the same time, just prior to, or just after the administration of a second compound described herein.
  • the term “therapeutic target” refers to a gene or gene product that, upon modulation of its activity (e.g., by modulation of expression, biological activity, and the like), can provide for modulation of the disease phenotype.
  • modulation is meant to refer to an increase or a decrease in the indicated phenomenon (e.g., modulation of a biological activity refers to an increase in a biological activity or a decrease in a biological activity).
  • standard of care refers to the treatment that is accepted by medical practitioners to be an appropriate, proper, effective, and/or widely used treatment for a certain disease.
  • the standard of care of a certain disease depends on many different factors, including the biological effect of treatment, region or location within the body, patient status (e.g. age, weight, gender, hereditary risks, other disabilities, secondary conditions), toxicity, metabolism, bioaccumulation, therapeutic index, dosage, and other factors known in the art.
  • Determining a standard of care for a disease is also dependent on establishing safety and efficacy in clinical trials as standardized by regulatory bodies such as the US Food and Drug Administration, International Council for Harmonisation, Health Canada, European Medicines Agency, Therapeutics Goods Administration, Central Drugs Standard Control Organization, National Medical Products Administration, Pharmaceuticals and Medical Devices Agency, Ministry of Food and Drug Safety, and the World Health Organization.
  • the standard of care for a disease may include but is not limited to surgery, radiation, chemotherapy, targeted therapy, or immunotherapy.
  • % w/w or “% wt/wt” means a percentage expressed in terms of the weight of the ingredient or agent over the total weight of the composition multiplied by 100.
  • CDRs complementarity determining regions
  • MSLN binding polypeptides comprise an immunoglobulin heavy chain variable domain comprising a CDR-H1, CDR-H2, and CDR-H3.
  • the CDR-H1 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 1-41.
  • CDR-H2 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 42-82.
  • the CDR-H3 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 83-123.
  • the CDR-H1 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 1-41;
  • the CDR-H2 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 42-82;
  • the CDR-H3 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 83-123.
  • the CDR-H1 comprises the sequence of SEQ ID NO: 1
  • the CDR-H2 comprises the sequence of SEQ ID NO: 42
  • the CDR-H3 comprises the sequence of SEQ ID NO: 83
  • the CDR-H1 comprises the sequence of SEQ ID NO: 2
  • the CDR-H2 comprises the sequence of SEQ ID NO: 43
  • the CDR-H3 comprises the sequence of SEQ ID NO: 84
  • the CDR-H1 comprises the sequence of SEQ ID NO: 3
  • the CDR-H2 comprises the sequence of SEQ ID NO: 44
  • the CDR-H3 comprises the sequence of SEQ ID NO: 85
  • the CDR-H1 comprises the sequence of SEQ ID NO: 4
  • the CDR-H2 comprises the sequence of SEQ ID NO: 45
  • the CDR-H3 comprises the sequence of SEQ ID NO: 86
  • the CDR-H1 comprises the sequence of SEQ ID NO: 5
  • the CDR-H2 comprises the sequence of SEQ ID NO: 83
  • the MSLN binding polypeptide comprise an immunoglobulin heavy chain variable domain comprising a CDR-H1, CDR-H2, and CDR-H3, where one or more of these CDRs are defined by a consensus sequence.
  • the consensus sequences provided herein have been derived from the alignments of CDRs depicted in FIG. 1 .
  • alternative alignments may be done (e.g. using global or local alignment, or with different algorithms, such as Hidden Markov Models, seeded guide trees, Needleman-Wunsch algorithm, or Smith-Waterman algorithm, or other known methods) and as such, alternative consensus sequences can be derived (including those done with a subset of the sequences provided herein).
  • the CDR-H1 is defined by the formula X 1 X 2 X 3 X 4 X 5 X 6 X 7 X 8 , where X 1 is G; X 2 is F, G, R, S, V, or Y; X 3 is I or T; X 4 is F, L, S, or Y; X 5 is D, G, N, R, or S; X 6 is A, F, H, I, L, S, V, or Y; X 7 is D, N, or Y; X 8 is A, F, L, S, T, V, or Y.
  • the CDR-H1 comprises a sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity to this consensus sequence. In some embodiments, the CDR-H1 comprises a sequence having 0, 1, 2, 3, 4, 5, or 6 substitutions from this consensus sequence.
  • the CDR-H2 is defined by the formula X 1 X 2 X 3 X 4 X 5 X 6 X 7 X 8 X 9 , where X 1 is no amino acid, S, or T; X 2 is I; X 3 is N, S, or, T; X 4 is A, P, R, S, or T; X 5 is F, H, I, L, T, V, or Y; X 6 is D, G, N, or S; X 7 is D, G, or S; X 8 is I, N, S, or T; X 9 is T.
  • the CDR-H2 comprises a sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity to this consensus sequence. In some embodiments, the CDR-H2 comprises a sequence having 0, 1, 2, 3, 4, 5, or 6 substitutions from this consensus sequence.
  • the CDR-H3 is defined by the formula X 1 X 2 X 3 X 4 X 5 X 6 X 7 X 8 X 9 X 10 X 11 X 12 X 13 X 14 X 15 X 16 X 17 X 18 X 19 X 20 X 21 , where X 1 is no amino acid or A; X 2 is no amino acid or L; X 3 is no amino acid or T; X 4 is no amino acid or R; X 5 is no amino acid or S; X 6 is no amino acid or D; X 7 is no amino acid or R; X 8 is no amino acid or P; X 9 is no amino acid, F, or V; X 10 is no amino acid, I, or T; X 11 is no amino acid, A, G, I, or V; X 12 is A, D, S, or V; X 13 is F, I, K, L, M, R, T, or Y; X 14 is A, E, F, H, I, K
  • the CDR-H3 comprises a sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity to this consensus sequence. In some embodiments, the CDR-H3 comprises a sequence having 0, 1, 2, 3, 4, 5, or 6 substitutions from this consensus sequence.
  • the heavy chain variable domain comprises an amino acid sequence having at least 90%, 95%, 99%, or 100% sequence identity to any sequence selected from SEQ ID NOs: 124-164.
  • the MSLN binding polypeptide is humanized. In some embodiments, the MSLN binding polypeptide is a single domain antibody (sdAb).
  • the MSLN binding polypeptide binds to MSLN with a dissociation constant (KD) of less than 1 nM, 2 nM, 5 nM, 10 nM, 15 nM, 20 nM, 30 nM, 40 nM, 50 nM, 60 nM, 70 nM, 80 nM, 90 nM, 100 nM, 200 nM, 300 nM, 400 nM, 500 nM, 600 nM, 700 nM, 800 nM, 900 nM, or 1000 nM, or any KD within a range defined by any two of the aforementioned KD.
  • KD dissociation constant
  • the binding polypeptides disclosed herein may be obtained from an antibody library.
  • the antibody library is an immune antibody library, a na ⁇ ve antibody library, a synthetic antibody library, or a semi-synthetic antibody library.
  • the antibody library comprises antibodies derived from human, or antibodies that are not immunogenic in humans, or both.
  • the antibody library comprises antibodies that are humanized, e.g. from mouse, rat, guinea pig, rabbit, cat, dog, cow, horse, sheep, goat, horse, donkey.
  • the antibody library comprises single domain antibodies (sdAb), nanobodies, V H H fragments, V NAR fragments, single-chain variable fragments (scFv), camelid antibodies, or cartilaginous fish antibodies, or any combination thereof.
  • sdAb single domain antibodies
  • nanobodies V H H fragments, V NAR fragments, single-chain variable fragments (scFv), camelid antibodies, or cartilaginous fish antibodies, or any combination thereof.
  • scFv single-chain variable fragments
  • camelid antibodies or cartilaginous fish antibodies, or any combination thereof.
  • sdAb single domain antibodies
  • scFv single-chain variable fragments
  • the antibody library comprises at least 100, 500, 1000, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 500000, or 1000000 unique antibodies, or any number of antibodies within a range defined by any two of the aforementioned number of antibodies.
  • Antibody libraries may be generated computationally or using machine learning processes.
  • An exemplary method of generating an antibody library computationally includes modifying a universal V H H framework with synthetic diversity in one or more complementary determining regions (CDRs), such as CDR1, CDR2, or CDR3, or any combination thereof.
  • CDRs complementary determining regions
  • the diversity of the CDRs are introduced by randomizing the library of sequences encoding for the antibodies with degenerate codons.
  • an NNK codon library can be employed, where the NNK codon comprises N (25% mix of A/T/C/G) and K (50% mix of T/G).
  • the NNK codon library is constructed with all possible amino acids, or with some amino acids (e.g. cysteine) and stop codon combinations excluded.
  • the antibody library can be generated using a trimer codon mix, which improves balanced representation of sense codons while reducing the chance of stop codons, improving efficiency of antibody generation and testing.
  • artificial intelligence-based prediction can be used to randomize specific binding pockets of the antibodies using available binding models or structure data.
  • panning the antibody library comprises screening for the candidate binding polypeptides by phage display, yeast display, bacterial display, ribosome display, or mRNA display, or any combination thereof.
  • panning the antibody library comprises one or more rounds of selection, wherein the candidate binding polypeptides are selected for specificity towards a cancer-associated antigen (e.g. MSLN) or cells or tissues displaying the cancer-associated antigen.
  • a cancer-associated antigen e.g. MSLN
  • the candidate binding polypeptides are selected under conditions including but not limited to tumor microenvironment-like conditions, immunosuppressive conditions, low or high pH, low or high oxygen concentrations, low or high temperatures, low or high viscosity, or any combination thereof, or for specificity towards modified or derivative forms of the one or more cancer-associated antigens.
  • the immunosuppressive conditions may comprise the presence of tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), tumor-associated neutrophils (TANs), cancer-associated fibroblasts (CAFs), or other immunosuppressive cells, or the presence of adenosine, or both.
  • the chimeric antigen receptor cells are from a cell line (e.g. Jurkat). In some embodiments, the chimeric antigen receptor cells are derived from a subject. In some embodiments, the subject has a cancer. In some embodiments, the subject has a cancer, and that cancer expresses any one or more of the cancer-associated antigens disclosed herein (e.g., MSLN). In some embodiments, the cancer is breast cancer, colorectal cancer, kidney cancer, liver cancer, lung cancer, brain cancer, pancreatic cancer, bladder cancer, testicular cancer, prostate cancer, gastric cancer, ovarian cancer, head and neck cancer, gallbladder cancer, a hematologic malignancy, or any combination thereof.
  • MSLN cancer-associated antigens disclosed herein
  • the cancer is breast cancer, colorectal cancer, kidney cancer, liver cancer, lung cancer, brain cancer, pancreatic cancer, bladder cancer, testicular cancer, prostate cancer, gastric cancer, ovarian cancer, head and neck cancer, gallbladder cancer,
  • the hematologic malignancy may comprise leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, hairy cell leukemia, lymphoma, Hodgkin's disease, Non-Hodgkin lymphoma, or multiple myeloma.
  • the subject is a mammal, such as a human, cat, dog, mouse, rat, hamster, rodent, cow, pig, horse, goat, sheep, donkey, or monkey. In some embodiments, the subject is a human.
  • CARs chimeric antigen receptors
  • the CAR comprises at least two binding polypeptides and the CAR is a multivalent CAR. In some embodiments, the CAR comprises two binding polypeptides and the CAR is a bivalent CAR. In some embodiments, the CAR comprises three binding polypeptides and the CAR is a trivalent CAR.
  • the CAR further comprises one or more signal peptides, linkers with various lengths and composition, hinges, transmembrane domains, costimulatory domains, signaling domains, cytoplasmic domains, functionality signals, proliferation signals, anti-exhaustion signals, anti-inhibitory receptors, tumor/cancer homing proteins, or regulatory molecules, or any combination thereof.
  • the hinges comprise CD3 ⁇ , CD4, CD8 or CD28 hinges, or computationally designed synthetic hinges with various lengths.
  • the transmembrane domains comprise CD3 ⁇ , CD4, CD8 or CD28 transmembrane domains, or computationally designed synthetic transmembrane domains.
  • the costimulatory domains comprise CD8, CD28, ICOS, 4-1BB, OX40 (CD134), CD27, CD40, CD40L, TLR or other TNFR superfamily member or Ig superfamily member costimulatory domains, or other signaling via cytoplasmic domains of IL-2R ⁇ , IL-15R- ⁇ , MyD88, or CD40 or any other Toll-like receptor or IL-1 receptor signaling pathway members.
  • the CARs disclosed herein are constructed by assembling CAR expression constructs from nucleic acids encoding for any one of the binding polypeptides disclosed herein and a mixture of compatible nucleic acids encoding for different CAR modules.
  • different combinations of CARs are produced for use in a CAR library for screening for CAR efficacy (in vitro or in vivo).
  • unique CARs are produced separately.
  • the CARs are specific for one target.
  • the CARs are specific for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 targets.
  • the CARs are bi-specific or tri-specific.
  • the nucleic acids encoding for the binding polypeptides identified by panning of the antibody library are assembled into CAR expression constructs with other CAR modules.
  • the CAR expression constructs are assembled using multi-fragment assembly reactions known in the art.
  • One exemplary method of assembling CAR expression constructs involves using Type IIS restriction enzymes to generate nucleic acid fragments with compatible overhang sequences and ligating the nucleic acid fragments with a ligase. As Type IIS restriction enzymes cleave outside of their recognition sites, multiple compatible nucleic acid fragments may be prepared simultaneously.
  • the CAR expression constructs can be assembled by overlap extension PCR.
  • the different CAR modules comprise signal peptides, linkers, hinges, transmembrane domains, costimulatory domains, activation domains, signaling domains, cytoplasmic domains, functionality signals, proliferation signals, anti-exhaustion signals, anti-inhibitor receptors, cancer homing proteins, or regulatory molecules, or any combination thereof.
  • Some exemplary hinges comprise CD8 hinge, CD28 hinge, IgG1 hinge, or IgG4 hinge.
  • Some exemplary transmembrane domains comprise CD3 ⁇ transmembrane domain, CD8 ⁇ transmembrane domain, CD4 transmembrane domain, CD28 transmembrane domain, or ICOS transmembrane domain.
  • Some exemplary costimulatory domains comprise CD8 costimulatory domain, CD28 costimulatory domain, 4-1BB costimulatory domain, OX40 (CD134) costimulatory domain, ICOS costimulatory domain, CD27 costimulatory domain, CD40 costimulatory domain, CD40L costimulatory domain, TLR costimulatory domains, MYD88-CD40 costimulatory domain, or KIR2DS2 costimulatory domain.
  • the different CAR modules are derived from CD8, CD28, 4-1BB, CD3 ⁇ , or any combination thereof.
  • the CAR may also be modified with various additions, including but not limited to cytokines, chemokines, cytokine receptors, chemokine receptors, antigen receptors or ligands, antibodies, or enzymes.
  • CAR chimeric antigen receptor
  • the CAR cell is a CAR-T cell.
  • the CAR cell is derived from a subject or from a cell line.
  • the subject has a cancer.
  • the subject has a cancer, and that cancer expresses any one or more of the cancer-associated antigens disclosed herein (e.g., MSLN).
  • the cancer is breast cancer, colorectal cancer, kidney cancer, liver cancer, lung cancer, brain cancer, pancreatic cancer, bladder cancer, testicular cancer, prostate cancer, gastric cancer, ovarian cancer, head and neck cancer, gallbladder cancer, a hematologic malignancy, or any combination thereof.
  • the hematologic malignancy may comprise leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, hairy cell leukemia, lymphoma, Hodgkin's disease, Non-Hodgkin lymphoma, or multiple myeloma.
  • nucleic acids that encode for a polypeptide.
  • the polypeptide is a binding polypeptide.
  • the polypeptide is a single domain binding polypeptide.
  • the polypeptide is any one of the binding polypeptides disclosed herein.
  • the polypeptide comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to: any one or more of the MSLN binding polypeptides disclosed herein.
  • the polypeptide is any one of the CARs disclosed herein.
  • any one of the nucleic acids that encode for a binding polypeptide can be prepared by recombinant DNA technology, synthetic chemistry techniques, or a combination thereof.
  • sequences of nucleic acids encoding for the binding polypeptide may be cloned into an expression vector using standard molecular techniques known in the art. Sequences can be obtained from other vectors encoding the desired protein sequence, from PCR-generated fragments using respective template nucleic acids, or by assembly of synthetic oligonucleotides encoding the desired sequences.
  • the expression vector may be a CAR expression vector, in which it is provided to an immune cell so that it expresses the CAR.
  • the expression vector may be an expression vector suited for large scale antibody or binding polypeptide production, from which the peptide products can be isolated for further use.
  • binding polypeptides or CARs may be confirmed by nucleic acid or protein assays known in the art.
  • the presence of transcribed mRNA of binding polypeptides or CARs can be detected and/or quantified by conventional hybridization assays (e.g. Northern blot analysis), amplification procedures (e.g. RT-PCR), SAGE (U.S. Pat. No. 5,695,937), and array-based technologies (see e.g. U.S. Pat. Nos. 5,405,783, 5,412,087 and 5,445,934), using probes complementary to any region of a polynucleotide that encodes for the binding polypeptides or CARs.
  • binding polypeptides or CARs can also be determined by examining the expressed peptide.
  • a variety of techniques are available in the art for protein analysis. They include but are not limited to radioimmunoassays, ELISA (enzyme linked immunoradiometric assays), “sandwich” immunoassays, immunoradiometric assays, in situ immunoassays (using e.g., colloidal gold, enzyme or radioisotope labels), western blot analysis, immunoprecipitation assays, immunofluorescent assays, and SDS-PAGE.
  • the methods comprise administering a chimeric antigen receptor cell to the subject. In some embodiments, the methods comprise administering any one of the chimeric antigen receptor cells disclosed herein. In some embodiments, the chimeric antigen receptor cell expresses and/or comprises any one or more of the MSLN binding polypeptides disclosed herein. In some embodiments, the chimeric antigen receptor cell is a CAR-T cell. In some embodiments, the chimeric antigen receptor cell is derived from the subject and is autologous to the subject. In some embodiments, the chimeric antigen receptor cell is allogeneic to the subject.
  • the chimeric antigen receptor cell is from a cell line (e.g. Jurkat).
  • the subject is a mammal, such as a human, cat, dog, mouse, rat, hamster, rodent, cow, pig, horse, goat, sheep, donkey, or monkey.
  • the subject is a human.
  • the subject has a cancer, and that cancer expresses any one or more of the cancer-associated antigens disclosed herein (e.g., MSLN).
  • the cancer is breast cancer, colorectal cancer, kidney cancer, liver cancer, lung cancer, brain cancer, pancreatic cancer, bladder cancer, testicular cancer, prostate cancer, gastric cancer, ovarian cancer, head and neck cancer, gallbladder cancer, a hematologic malignancy, or any combination thereof.
  • the hematologic malignancy may comprise leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, hairy cell leukemia, lymphoma, Hodgkin's disease, Non-Hodgkin lymphoma, or multiple myeloma.
  • the chimeric antigen receptor cell is administered parenterally.
  • the chimeric antigen receptor cell is administered once per day, twice per day, three times per day or more. In some embodiments, the chimeric antigen receptor cell is administered daily, every day, every alternate day, five days a week, once a week, every other week, two weeks per month, three weeks per month, once a month, twice a month, three times per month, or more. In some embodiments, the immune cell is administered for at least 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, 18 months, 2 years, 3 years, or more.
  • the amount of a given agent that correspond to such an amount varies depending upon factors such as the particular compound, the severity of the disease, the identity (e.g., weight) of the subject or host in need of treatment, but nevertheless is routinely determined in a manner known in the art according to the particular circumstances surrounding the case, including, e.g., the specific agent being administered, the route of administration, and the subject or host being treated.
  • the desired dose is conveniently presented in a single dose or as divided doses administered simultaneously (or over a short period of time) or at appropriate intervals, for example as two, three, four or more sub-doses per day.
  • toxicity and therapeutic efficacy of such therapeutic regimens are determined by standard pharmaceutical procedures in cell cultures or experimental animals, including, but not limited to, the determination of the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between the toxic and therapeutic effects is the therapeutic index and it is expressed as the ratio between LD50 and ED50.
  • Compounds exhibiting high therapeutic indices are preferred.
  • the data obtained from cell culture assays and animal studies are used in formulating a range of dosage for use in human.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with minimal toxicity. The dosage varies within this range depending upon the dosage form employed and the route of administration utilized.
  • the Maxi Plasmid Purification kit (Zymo, D4203) was used for CAR plasmid preparation. Plasmid concentration and quality was analyzed by Nanodrop (260/280 ratio and 260/230 ratio) and the ToxinSensor Chromogenic LAL Endotoxin Assay (Genescript, L00350). Good-quality plasmid DNA will have an A260/A280 ratio of 1.8-2.0, an A260/A230 ratio greater than 2.0, and less than 0.1 EU/ ⁇ g of endotoxin.
  • the CAR constructs contain a signal peptide (e.g. CD8 signal peptide), hinge (e.g. CD8 ⁇ stalk), transmembrane domain (e.g. CD8 transmembrane domain or CD28 transmembrane domain), and signaling domains (e.g. 4-1BB costimulatory domain or CD3 ⁇ signaling domain) and the anti-MSLN sdAb.
  • signal peptide e.g. CD8 signal peptide
  • hinge e.g. CD8 ⁇ stalk
  • transmembrane domain e.g. CD8 transmembrane domain or CD28 transmembrane domain
  • signaling domains e.g. 4-1BB costimulatory domain or CD3 ⁇ signaling domain
  • the expressed cassette of the CAR plasmids can also be engineered to express fluorescent eGFP+T2A self-cleaving peptide sequences.
  • truncated CD19 or truncated EGFR cassettes can be used to monitor by antibody detection.
  • Human sequences e.g. CD8, 4-1BB, CD3 ⁇
  • GenBank GenBank
  • human embryonic kidney 293T (HEK293T) cells were co-transfected with CAR transgene-encoding pLenti transfer plasmid and one or more necessary packaging plasmids (i.e. encoding for Gag, Pol, Rev, VSVG, and optionally Tat).
  • the supernatants of the HEK293T cultures were collected at either 48 or 72 hours after transfection, centrifuged, and filtered with a 0.45 ⁇ m filter.
  • Virus titration was done in Jurkat cells transduced with diluted lentivirus collections. After 48 hours, transduced Jurkat cells were stained with biotinylated recombinant protein L and phycoerythrin (PE)-conjugated streptavidin, and anti-MSLN CAR abundance was measured by flow cytometry.
  • PE phycoerythrin
  • Human PBMCs were isolated from peripheral blood of healthy human donors by density gradient centrifugation with the Lymphoprep reagent (StemCell Technologies). PBMCs were resuspended at 1 ⁇ 10 6 cells/mL in X-VIVO 15 serum-free hematopoietic medium (Lonza, 04-418QCN) with 10 ng/mL IL-2 (Novoprotein, GMP-CD66) and 10 ng/mL IL-7 (Novoprotein, GMP-CD47). PBMCs were stimulated with 50 ng/mL anti-CD3 antibody (Novoprotein, GMP-A018) for 24 hours.
  • PBMCs were transduced with anti-MSLN CAR lentivirus (at MOI of 1-10) and expanded in appropriate flasks for 9-10 days in RPMI 1640 basal medium supplemented with 10% fetal bovine serum. T cells were rested for 24 hours at 37° C. before any assay.
  • CAR surface levels, CD3, and CD4/CD8 ratios were measured 12 or 14 days after initial stimulation of PBMCs with the anti-CD3 antibody.
  • CAR-T expansion After 14 days of CAR-T expansion, anti-MSLN CAR-T cells were centrifuged and the supernatant was discarded. The cell pellet was resuspended in chilled CryoStor CS10 (StemCell Technologies, 07930) at a viable cell density of 5 ⁇ 10 7 cells/mL. Aliquots of cell suspension were dispensed into cryovials. The cryovials were cooled with a 1° C./minute decrease. The frozen cells were transferred to liquid nitrogen.
  • the cells are quickly thawed in a 37° C. water bath with gentle agitation.
  • the thawed cells are transferred to a 50 mL conical tube and washed by adding 20 mL of fresh growth medium dropwise.
  • the cells are centrifuged and resuspended in X-VIVO 15 medium at a cell density of 1 ⁇ 10 6 cells/mL.
  • cytotoxicity of anti-MSLN CAR T-cells was determined by standard luciferase-based assays. Briefly, target cells expressing firefly luciferase were co-cultured with CAR-T cells in triplicate at the indicated effector:target ratios using white-walled 96-well plates with 2 ⁇ 10 4 target cells in a total volume of 100 ⁇ L per well in X-VIVO 15 medium. Target cells alone as a control were plated at the same cell density. After 48 hours of co-culture, 100 ⁇ L of luciferase substrate (ONE-Glo, Promega) was directly added to each well. Emitted light was detected with a luminescence plate reader.
  • ONE-Glo ONE-Glo, Promega
  • target cancer cells engineered to overexpress luciferase were prepared in a cell culture medium (such as RPMI) and seeded on to 96 well plates with 5 ⁇ 10 4 to 5 ⁇ 10 6 cells in a volume of 100 ⁇ L/well, and incubated at 37° C. for 24 hours. Then, CAR T-cells were seeded in corresponding wells with CAR T-cells at various effector:target (E:T) ratios (e.g., 5:1, 1:1, and 1:5) in 100 ⁇ L/well. The co-cultures were then incubated at 37° C. for 24 or 48 hours.
  • E:T effector:target
  • Interferon gamma (IFN- ⁇ ) and IL-2 secretion was quantitated through enzyme-linked immunosorbent assay (ELISA) using the human IL-2 ELISA Kit II (BD, 550611) and human IFN- ⁇ ELISA Kit II (BD, 550612)
  • CAR T-cells 1 ⁇ 10 6 per well of anti-MSLN CAR T-cells were stimulated with target cells at indicated effector:target cell ratios in 6-well tissue culture plates for 24 hours.
  • Cells were stained with LIVE/DEAD Fixable Near-IR Dead Cell Stain Kit (ThermoFisher) to label dead cells and with anti-CD3, and CAR-T cells were identified as CD3+GFP+cells by flow cytometry.
  • CD69 and CD25 on CAR-T cells were stained with Alexa Fluor 700 anti-human CD69 antibody (Biolegend) and PE anti-CD25 antibody (Biolegend).
  • Genomic DNA was extracted using the Blood/Cell Genome DNA Mini kit (Tiangen, DP304-03). 75 ng of gDNA was used for PCT to amplify the CAR region of the library. The amplified region was TA cloned with the TA/Blunt-Zero Cloning kit (Vazyme, C601) and sequenced.
  • anti-MSLN CAR T-cell lines were prepared from the anti-MSLN binders disclosed herein. Constructs were expressed in T-cells isolated from two donors, denoted Z0016 and Y1287. Cytotoxicity assays were performed as described in Example 1, at effector:target ratios of 1:5, 1:1, and 5:1. These anti-MSLN CAR T-cell lines showed cytotoxic efficacy against BxPC-3 pancreatic cancer cells ( FIG. 2 A ), Capan-1 pancreatic cancer cells ( FIG. 2 B ), and NCI-N87 (N87) gastric cancer cells ( FIG. 2 C ). Untransfected T-cells (UNT) were used as negative control.
  • An exemplary anti-MSLN CAR T-cell line B4T2-002 was tested in in vivo NSG mouse models.
  • administration of B4T2-002 resulted in dramatic clearance of the xenograft in HCC1806 breast cancer, H2110 non-small cell lung cancer, and N87 gastric cancer models, whereas tumor growth in control mice progressed ( FIG. 3 ).
  • a patient presents with a cancer, for example, a cancer that expresses or overexpresses MSLN (e.g., such that MSLN can be used as a biomarker to detect and target the cancer).
  • a cancer for example, a cancer that expresses or overexpresses MSLN (e.g., such that MSLN can be used as a biomarker to detect and target the cancer).
  • the cancer is breast cancer, colorectal cancer, kidney cancer, liver cancer, lung cancer, brain cancer, pancreatic cancer, bladder cancer, testicular cancer, prostate cancer, gastric cancer, ovarian cancer, head and neck cancer, gallbladder cancer, a hematologic malignancy, or any combination thereof.
  • the hematologic malignancy may comprise leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, hairy cell leukemia, lymphoma, Hodgkin's disease, Non-Hodgkin lymphoma, or multiple myeloma.
  • One or more of the CAR T-cells disclosed herein are administered to the patient enterally, orally, intranasally, parenterally, intravenously, intraperitoneally, intramuscularly, intra-arterially, intraventricularly, intradermally, intralesionally, intracranially, intrathecally, or subcutaneously.
  • the CAR T-cells are administered as doses in the amount of 10 4 , 10 5 , 10 6 , 10 7 , 10 8 , or 10 9 cells per dose, or any number of cells within a range defined by any two of the aforementioned cells per dose, or any number of cells that is effective and/or safe as determined by a trained medical practitioner.
  • the doses are administered every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 24, 36, or 48 days, or weeks, or any time within a range defined by any two of the aforementioned times, or any timing of dosing that is effective and/or safe as determined by a trained medical practitioner.
  • An improvement of the cancer or symptoms thereof is observed in the patient following administration of the one or more CAR T-cells.
  • Administration of the CAR T-cells may be performed in conjunction with another therapy for cancer, including but not limited to immunotherapy, chemotherapy, radiation therapy, surgery, photodynamic therapy, or targeted therapy.
  • a range includes each individual member.
  • a group having 1-3 articles refers to groups having 1, 2, or 3 articles.
  • a group having 1-5 articles refers to groups having 1, 2, 3, 4, or 5 articles, and so forth.

Abstract

Disclosed herein are mesothelin (MSLN)-specific binding polypeptides. These binding polypeptides may be incorporated into chimeric antigen receptors (CARs). Also disclosed herein are methods of using these binding polypeptides and/or CARs for the treatment of, for example, a cancer.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit of priority of U.S. Provisional Patent Application No. 63/127,885, filed Dec. 18, 2020, and U.S. Provisional Patent Application No. 63/262,309, filed Oct. 8, 2021, each of which is hereby expressly incorporated by reference in its entirety.
  • REFERENCE TO SEQUENCE LISTING
  • The present application is being filed along with a Sequence Listing in electronic format. The Sequence Listing is provided as a file entitled BWGB006_SeqListing.TXT, which was created and last modified on Dec. 16, 2021, which is 69,451 bytes in size. The information in the electronic Sequence Listing is hereby incorporated by reference in its entirety.
  • FIELD
  • Aspects of the present disclosure relate generally to mesothelin (MSLN)-specific binding polypeptides. These binding polypeptides can be incorporated into chimeric antigen receptor (CAR) constructs to be expressed in immune cells. These binding polypeptides and CARs may be used in the treatment of cancer.
  • BACKGROUND
  • Chimeric antigen receptor (CAR) T cells and other adoptive cell therapies have been shown to be effective in the treatment of cancer. The CAR, which is made up of an extracellular antigen binding domain, a transmembrane domain, and an intracellular signaling domain, enables directed killing of cancer cells based on cell surface antigen expression while minimally affecting normal cells that are not expressing the targeted antigen. The extracellular antigen binding domain is often made up of an antibody or a binding fragment or derivative thereof, such as a single chain variable fragment (scFv) or single domain antibody (sdAb). There is a present need for improved extracellular antigen binding domains to be used in CARs for the treatment of various cancers or other diseases.
  • SUMMARY OF THE DISCLOSURE
  • Disclosed herein are binding polypeptides that are able to bind to mesothelin (MSLN). These binding polypeptides may be incorporated in a chimeric antigen receptor (CAR), which can be expressed by a cell. In some embodiments, the binding polypeptides are single domain antibodies (sdAbs).
  • Disclosed herein in some embodiments are MSLN binding polypeptides comprising an immunoglobulin heavy chain variable domain comprising a CDR-H1, CDR-H2, and CDR-H3. In some embodiments, the CDR-H1 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 1-41. In some embodiments, CDR-H2 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 42-82. In some embodiments, the CDR-H3 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 83-123. In some embodiments, the CDR-H1 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 1-41, the CDR-H2 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 42-82, and the CDR-H3 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 83-123. In some embodiments, the immunoglobulin heavy chain variable domain comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 124-164.
  • Also disclosed herein are nucleic acids that encode for any one of the MSLN binding polypeptides disclosed herein.
  • Also disclosed herein are methods of treating a cancer in a subject in need thereof. In some embodiments, the methods comprise administering a chimeric antigen receptor cell to the subject. In some embodiments, the chimeric antigen receptor cell is any one of the chimeric antigen receptor cells disclosed herein. In some embodiments, the chimeric antigen receptor cell comprises any one or more of the MSLN binding polypeptides disclosed herein.
  • Embodiments of the present invention provided herein are described by way of the following numbered alternatives:
  • 1. A mesothelin (MSLN) binding polypeptide comprising an immunoglobulin heavy chain variable domain comprising a CDR-H1, CDR-H2, and CDR-H3, wherein:
      • the CDR-H1 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 1-41;
      • the CDR-H2 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 42-82; and
      • the CDR-H3 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 83-123.
  • 2. The MSLN binding polypeptide of alternative 1, wherein:
      • 1) the CDR-H1 comprises the sequence of SEQ ID NO: 1, the CDR-H2 comprises the sequence of SEQ ID NO: 42, and the CDR-H3 comprises the sequence of SEQ ID NO: 83;
      • 2) the CDR-H1 comprises the sequence of SEQ ID NO: 2, the CDR-H2 comprises the sequence of SEQ ID NO: 43, and the CDR-H3 comprises the sequence of SEQ ID NO: 84;
      • 3) the CDR-H1 comprises the sequence of SEQ ID NO: 3, the CDR-H2 comprises the sequence of SEQ ID NO: 44, and the CDR-H3 comprises the sequence of SEQ ID NO: 85;
      • 4) the CDR-H1 comprises the sequence of SEQ ID NO: 4, the CDR-H2 comprises the sequence of SEQ ID NO: 45, and the CDR-H3 comprises the sequence of SEQ ID NO: 86;
      • 5) the CDR-H1 comprises the sequence of SEQ ID NO: 5, the CDR-H2 comprises the sequence of SEQ ID NO: 46, and the CDR-H3 comprises the sequence of SEQ ID NO: 87;
      • 6) the CDR-H1 comprises the sequence of SEQ ID NO: 6, the CDR-H2 comprises the sequence of SEQ ID NO: 47, and the CDR-H3 comprises the sequence of SEQ ID NO: 88;
      • 7) the CDR-H1 comprises the sequence of SEQ ID NO: 7, the CDR-H2 comprises the sequence of SEQ ID NO: 48, and the CDR-H3 comprises the sequence of SEQ ID NO: 89;
      • 8) the CDR-H1 comprises the sequence of SEQ ID NO: 8, the CDR-H2 comprises the sequence of SEQ ID NO: 49, and the CDR-H3 comprises the sequence of SEQ ID NO: 90;
      • 9) the CDR-H1 comprises the sequence of SEQ ID NO: 9, the CDR-H2 comprises the sequence of SEQ ID NO: 50, and the CDR-H3 comprises the sequence of SEQ ID NO: 91;
      • 10) the CDR-H1 comprises the sequence of SEQ ID NO: 10, the CDR-H2 comprises the sequence of SEQ ID NO: 51, and the CDR-H3 comprises the sequence of SEQ ID NO: 92;
      • 11) the CDR-H1 comprises the sequence of SEQ ID NO: 11, the CDR-H2 comprises the sequence of SEQ ID NO: 52, and the CDR-H3 comprises the sequence of SEQ ID NO: 93;
      • 12) the CDR-H1 comprises the sequence of SEQ ID NO: 12, the CDR-H2 comprises the sequence of SEQ ID NO: 53, and the CDR-H3 comprises the sequence of SEQ ID NO: 94;
      • 13) the CDR-H1 comprises the sequence of SEQ ID NO: 13, the CDR-H2 comprises the sequence of SEQ ID NO: 54, and the CDR-H3 comprises the sequence of SEQ ID NO: 95;
      • 14) the CDR-H1 comprises the sequence of SEQ ID NO: 14, the CDR-H2 comprises the sequence of SEQ ID NO: 55, and the CDR-H3 comprises the sequence of SEQ ID NO: 96;
      • 15) the CDR-H1 comprises the sequence of SEQ ID NO: 15, the CDR-H2 comprises the sequence of SEQ ID NO: 56, and the CDR-H3 comprises the sequence of SEQ ID NO: 97;
      • 16) the CDR-H1 comprises the sequence of SEQ ID NO: 16, the CDR-H2 comprises the sequence of SEQ ID NO: 57, and the CDR-H3 comprises the sequence of SEQ ID NO: 98;
      • 17) the CDR-H1 comprises the sequence of SEQ ID NO: 17, the CDR-H2 comprises the sequence of SEQ ID NO: 58, and the CDR-H3 comprises the sequence of SEQ ID NO: 99;
      • 18) the CDR-H1 comprises the sequence of SEQ ID NO: 18, the CDR-H2 comprises the sequence of SEQ ID NO: 59, and the CDR-H3 comprises the sequence of SEQ ID NO: 100;
      • 19) the CDR-H1 comprises the sequence of SEQ ID NO: 19, the CDR-H2 comprises the sequence of SEQ ID NO: 60, and the CDR-H3 comprises the sequence of SEQ ID NO: 101;
      • 20) the CDR-H1 comprises the sequence of SEQ ID NO: 20, the CDR-H2 comprises the sequence of SEQ ID NO: 61, and the CDR-H3 comprises the sequence of SEQ ID NO: 102;
      • 21) the CDR-H1 comprises the sequence of SEQ ID NO: 21, the CDR-H2 comprises the sequence of SEQ ID NO: 62, and the CDR-H3 comprises the sequence of SEQ ID NO: 103;
      • 22) the CDR-H1 comprises the sequence of SEQ ID NO: 22, the CDR-H2 comprises the sequence of SEQ ID NO: 63, and the CDR-H3 comprises the sequence of SEQ ID NO: 104;
      • 23) the CDR-H1 comprises the sequence of SEQ ID NO: 23, the CDR-H2 comprises the sequence of SEQ ID NO: 64, and the CDR-H3 comprises the sequence of SEQ ID NO: 105;
      • 24) the CDR-H1 comprises the sequence of SEQ ID NO: 24, the CDR-H2 comprises the sequence of SEQ ID NO: 65, and the CDR-H3 comprises the sequence of SEQ ID NO: 106;
      • 25) the CDR-H1 comprises the sequence of SEQ ID NO: 25, the CDR-H2 comprises the sequence of SEQ ID NO: 66, and the CDR-H3 comprises the sequence of SEQ ID NO: 107;
      • 26) the CDR-H1 comprises the sequence of SEQ ID NO: 26, the CDR-H2 comprises the sequence of SEQ ID NO: 67, and the CDR-H3 comprises the sequence of SEQ ID NO: 108;
      • 27) the CDR-H1 comprises the sequence of SEQ ID NO: 27, the CDR-H2 comprises the sequence of SEQ ID NO: 68, and the CDR-H3 comprises the sequence of SEQ ID NO: 109;
      • 28) the CDR-H1 comprises the sequence of SEQ ID NO: 28, the CDR-H2 comprises the sequence of SEQ ID NO: 69, and the CDR-H3 comprises the sequence of SEQ ID NO: 110;
      • 29) the CDR-H1 comprises the sequence of SEQ ID NO: 29, the CDR-H2 comprises the sequence of SEQ ID NO: 70, and the CDR-H3 comprises the sequence of SEQ ID NO: 111;
      • 30) the CDR-H1 comprises the sequence of SEQ ID NO: 30, the CDR-H2 comprises the sequence of SEQ ID NO: 71, and the CDR-H3 comprises the sequence of SEQ ID NO: 112;
      • 31) the CDR-H1 comprises the sequence of SEQ ID NO: 31, the CDR-H2 comprises the sequence of SEQ ID NO: 72, and the CDR-H3 comprises the sequence of SEQ ID NO: 113;
      • 32) the CDR-H1 comprises the sequence of SEQ ID NO: 32, the CDR-H2 comprises the sequence of SEQ ID NO: 73, and the CDR-H3 comprises the sequence of SEQ ID NO: 114;
      • 33) the CDR-H1 comprises the sequence of SEQ ID NO: 33, the CDR-H2 comprises the sequence of SEQ ID NO: 74, and the CDR-H3 comprises the sequence of SEQ ID NO: 115;
      • 34) the CDR-H1 comprises the sequence of SEQ ID NO: 34, the CDR-H2 comprises the sequence of SEQ ID NO: 75, and the CDR-H3 comprises the sequence of SEQ ID NO: 116;
      • 35) the CDR-H1 comprises the sequence of SEQ ID NO: 35, the CDR-H2 comprises the sequence of SEQ ID NO: 76, and the CDR-H3 comprises the sequence of SEQ ID NO: 117;
      • 36) the CDR-H1 comprises the sequence of SEQ ID NO: 36, the CDR-H2 comprises the sequence of SEQ ID NO: 77, and the CDR-H3 comprises the sequence of SEQ ID NO: 118;
      • 37) the CDR-H1 comprises the sequence of SEQ ID NO: 37, the CDR-H2 comprises the sequence of SEQ ID NO: 78, and the CDR-H3 comprises the sequence of SEQ ID NO: 119;
      • 38) the CDR-H1 comprises the sequence of SEQ ID NO: 38, the CDR-H2 comprises the sequence of SEQ ID NO: 79, and the CDR-H3 comprises the sequence of SEQ ID NO: 120;
      • 39) the CDR-H1 comprises the sequence of SEQ ID NO: 39, the CDR-H2 comprises the sequence of SEQ ID NO: 80, and the CDR-H3 comprises the sequence of SEQ ID NO: 121;
      • 40) the CDR-H1 comprises the sequence of SEQ ID NO: 40, the CDR-H2 comprises the sequence of SEQ ID NO: 81, and the CDR-H3 comprises the sequence of SEQ ID NO: 122; or
      • 41) the CDR-H1 comprises the sequence of SEQ ID NO: 41, the CDR-H2 comprises the sequence of SEQ ID NO: 82, and the CDR-H3 comprises the sequence of SEQ ID NO: 123.
  • 3. The MSLN binding polypeptide of alternative 1 or 2, wherein the heavy chain variable domain comprises an amino acid sequence having at least 90%, 95%, 99%, or 100% sequence identity to any sequence selected from SEQ ID NOs: 124-164.
  • 4. The MSLN binding polypeptide of any one of alternatives 1-3, wherein the MSLN binding polypeptide is humanized.
  • 5. The MSLN binding polypeptide of any one of alternatives 1-4, wherein the MSLN binding polypeptide is a single domain antibody (sdAb).
  • 6. A chimeric antigen receptor (CAR) comprising the MSLN binding polypeptide of any one of alternatives 1-5.
  • 7. A chimeric antigen receptor (CAR) cell comprising the CAR of alternative 6.
  • 8. The CAR cell of alternative 7, wherein the CAR cell is a CART cell.
  • 9. The CAR cell of alternative 7 or 8, wherein the CAR cell comprises at least two binding polypeptides and the CAR cell is a multivalent CAR cell.
  • 10. The CAR cell of any one of alternatives 7-9, wherein the CAR cell is derived from a subject or from a cell line.
  • 11. The CAR cell of alternative 10, wherein the subject has a cancer.
  • 12. The CAR cell of alternative 11, wherein the cancer is breast cancer, colorectal cancer, kidney cancer, liver cancer, lung cancer, brain cancer, pancreatic cancer, bladder cancer, testicular cancer, prostate cancer, gastric cancer, ovarian cancer, head and neck cancer, gallbladder cancer, a hematologic malignancy, or any combination thereof.
  • 13. A nucleic acid that encodes for a polypeptide comprising a sequence having at least 90%, 95%, 99%, or 100% sequence identity to the MSLN binding polypeptide of any one of alternatives 1-5 or the CAR of alternative 6.
  • 14. A method of treating a cancer in a subject in need thereof, comprising administering the CAR cell of any one of alternatives 7-12.
  • 15. The method of alternative 14, wherein the chimeric antigen receptor cell is autologous or allogeneic to the subject.
  • 16. The method of alternative 14 or 15, wherein the subject is a mammal.
  • 17. The method of any one of alternatives 14-16, wherein the subject is a human.
  • 18. The method of any one of alternatives 14-17, wherein the cancer is breast cancer, colorectal cancer, kidney cancer, liver cancer, lung cancer, brain cancer, pancreatic cancer, bladder cancer, testicular cancer, prostate cancer, gastric cancer, ovarian cancer, head and neck cancer, gallbladder cancer, a hematologic malignancy, or any combination thereof.
  • 19. The method of any one of alternatives 14-18, wherein the chimeric antigen receptor cell is administered parenterally.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • In addition to the features described above, additional features and variations will be readily apparent from the following descriptions of the drawings and exemplary embodiments. It is to be understood that these drawings depict typical embodiments and are not intended to be limiting in scope.
  • FIG. 1 depicts an exemplary alignment for the heavy chain variable domain CDRs disclosed herein.
  • FIG. 2A depicts a bar chart which shows the results of a cytotoxicity assay of exemplary anti-MSLN CAR T-cell lines against BxPC-3 pancreatic cancer cells.
  • FIG. 2B depicts a bar chart which shows the results of a cytotoxicity assay of exemplary anti-MSLN CAR T-cell lines against Capan-1 pancreatic cancer cells.
  • FIG. 2C depicts a bar chart which shows the results of a cytotoxicity assay of exemplary anti-MSLN CAR T-cell lines against NCI-N87 gastric cancer cells, where the T-cells have been obtained from a donor denoted Z0016.
  • FIG. 2D depicts a bar chart which shows the results of a cytotoxicity assay of exemplary anti-MSLN CAR T-cell lines against NCI-N87 gastric cancer cells, where the T-cell have been obtained from a donor denoted Y1287.
  • FIG. 3 depicts three line graphs which show the anti-tumor efficacy of an exemplary anti-MSLN CAR T-cell line B4T2-002 ('002) against HCC1806 breast cancer, H2110 non-small cell lung cancer, and NCI-N87 gastric cancer xenograph models.
  • DETAILED DESCRIPTION OF THE DISCLOSURE
  • Disclosed herein are binding polypeptides that are incorporated into a chimeric antigen receptor cell. In some embodiments, the chimeric antigen receptor cell is a chimeric antigen receptor T cell (CAR-T cell). These CAR-Ts may be constructed through processes conventionally known in the art. The binding polypeptides provide specificity towards their respective tumor-associated antigens, enabling targeting of cancers expressing said tumor-associated antigens by the CAR-T cell.
  • In some embodiments, the binding polypeptides are single domain antibodies (sdAbs) disposed on the surface of the chimeric antigen receptor cells (e.g. CAR-T cell). The sdAbs may be specific for, or have binding affinity towards, a tumor-associated antigen. In some embodiments, the tumor-associated antigen is mesothelin (MSLN).
  • Also disclosed herein are methods of treating a cancer in a subject in need thereof by administering a chimeric antigen receptor cell comprising one or more of the binding polypeptides disclosed herein. In some embodiments, the cancer may be breast cancer, colorectal cancer, kidney cancer, liver cancer, lung cancer, brain cancer, pancreatic cancer, bladder cancer, testicular cancer, prostate cancer, gastric cancer, ovarian cancer, head and neck cancer, gallbladder cancer, a hematologic malignancy, or any combination thereof. In some embodiments, the hematologic malignancy may comprise leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, hairy cell leukemia, lymphoma, Hodgkin's disease, Non-Hodgkin lymphoma, or multiple myeloma. The CAR-T cell may be derived from the subject for an autologous treatment. Alternatively, the CAR-T cell may be derived from the same species as the subject for an allogeneic treatment.
  • Definitions
  • Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which the claimed subject matter belongs. It is to be understood that the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of any subject matter claimed.
  • The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described.
  • The articles “a” and “an” are used herein to refer to one or to more than one (for example, at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element.
  • By “about” is meant a quantity, level, value, number, frequency, percentage, dimension, size, amount, weight or length that varies by as much as 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1% to a reference quantity, level, value, number, frequency, percentage, dimension, size, amount, weight or length.
  • Throughout this specification, unless the context requires otherwise, the words “comprise,” “comprises,” and “comprising” will be understood to imply the inclusion of a stated step or element or group of steps or elements but not the exclusion of any other step or element or group of steps or elements. By “consisting of” is meant including, and limited to, whatever follows the phrase “consisting of.” Thus, the phrase “consisting of” indicates that the listed elements are required or mandatory, and that no other elements may be present. By “consisting essentially of” is meant including any elements listed after the phrase and limited to other elements that do not interfere with or contribute to the activity or action specified in the disclosure for the listed elements. Thus, the phrase “consisting essentially of” indicates that the listed elements are required or mandatory, but that other elements are optional and may or may not be present depending upon whether or not they materially affect the activity or action of the listed elements.
  • As used herein, the terms “individual(s)”, “subject(s)” and “patient(s)” mean any mammal. In some embodiments, the mammal is a human. In some embodiments, the mammal is a non-human. None of the terms require or are limited to situations characterized by the supervision (e.g. constant or intermittent) of a health care worker (e.g. a doctor, a registered nurse, a nurse practitioner, a physician's assistant, an orderly or a hospice worker).
  • The term “administering” includes enteral, oral, intranasal, parenteral, intravenous, intraperitoneal, intramuscular, intra-arteriole, intraventricular, intradermal, intralesional, intracranial, intrathecal, or subcutaneous administration, or the implantation of a slow-release device, e.g., a mini-osmotic pump, to a subject.
  • The terms “nucleic acid” or “nucleic acid molecule” as used herein refers to polynucleotides, such as deoxyribonucleic acid (DNA) or ribonucleic acid (RNA), oligonucleotides, fragments generated by the polymerase chain reaction (PCR), and fragments generated by any of ligation, scission, endonuclease action, and exonuclease action. Nucleic acid molecules can be composed of monomers that are naturally-occurring nucleotides (such as DNA and RNA), or analogs of naturally-occurring nucleotides (e.g., enantiomeric forms of naturally-occurring nucleotides), or a combination of both. Nucleic acid monomers can be linked by phosphodiester bonds or analogs of such linkages. A nucleic acid or nucleic acids can be contained in a nucleic acid vector or nucleic acid construct (e.g. plasmid, virus, bacteriophage, cosmid, fosmid, phagemid, bacterial artificial chromosome (BAC), yeast artificial chromosome (YAC), or human artificial chromosome (HAC)) that can be used for amplification and/or expression of the nucleic acid or nucleic acids in various biological systems. Typically, the vector or construct will also contain elements including but not limited to promoters, enhancers, terminators, inducers, ribosome binding sites, translation initiation sites, start codons, stop codons, polyadenylation signals, origins of replication, cloning sites, multiple cloning sites, restriction enzyme sites, epitopes, reporter genes, selection markers, antibiotic selection markers, targeting sequences, peptide purification tags, or accessory genes, or any combination thereof.
  • A nucleic acid or nucleic acid molecule can comprise one or more sequences encoding different peptides, polypeptides, or proteins. These one or more sequences can be joined in the same nucleic acid or nucleic acid molecule adjacently, or with extra nucleic acids in between, e.g. linkers, repeats or restriction enzyme sites, or any other sequence that is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, or 300 bases long, or any length in a range defined by any two of the aforementioned lengths. The term “downstream” on a nucleic acid as used herein refers to a sequence being after the 3′-end of a previous sequence, on the strand containing the encoding sequence (sense strand) if the nucleic acid is double stranded. The term “upstream” on a nucleic acid as used herein refers to a sequence being before the 5′-end of a subsequent sequence, on the strand containing the encoding sequence (sense strand) if the nucleic acid is double stranded. The term “grouped” on a nucleic acid as used herein refers to two or more sequences that occur in proximity either directly or with extra nucleic acids in between, e.g. linkers, repeats, or restriction enzyme sites, or any other sequence that is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, or 300 bases long, or any length in a range defined by any two of the aforementioned lengths, but generally not with a sequence in between that encodes for a functioning or catalytic polypeptide, protein, or protein domain.
  • The term “codon optimized” regarding a nucleic acid as used herein refers to the substitution of codons of the nucleic acid to enhance or maximize translation in a host of a particular species without changing the polypeptide sequence based on species-specific codon usage biases and relative availability of each aminoacyl-tRNA in the target cell cytoplasm. Codon optimization and techniques to perform such optimization is known in the art. Those skilled in the art will appreciate that gene expression levels are dependent on many factors, such as promoter sequences and regulatory elements. In this aspect, many synthetic genes can be designed to increase their protein expression level.
  • The terms “peptide”, “polypeptide”, and “protein” as used herein refers to macromolecules comprised of amino acids linked by peptide bonds. The numerous functions of peptides, polypeptides, and proteins are known in the art, and include but are not limited to enzymes, structure, transport, defense, hormones, or signaling. Peptides, polypeptides, and proteins are often, but not always, produced biologically by a ribosomal complex using a nucleic acid template, although chemical syntheses are also available. By manipulating the nucleic acid template, peptide, polypeptide, and protein mutations such as substitutions, deletions, truncations, additions, duplications, or fusions of more than one peptide, polypeptide, or protein can be performed. These fusions of more than one peptide, polypeptide, or protein can be joined in the same molecule adjacently, or with extra amino acids in between, e.g. linkers, repeats, epitopes, or tags, or any other sequence that is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, or 300 bases long, or any length in a range defined by any two of the aforementioned lengths. The term “downstream” on a polypeptide as used herein refers to a sequence being after the C-terminus of a previous sequence. The term “upstream” on a polypeptide as used herein refers to a sequence being before the N-terminus of a subsequent sequence.
  • In some embodiments, the nucleic acid or peptide sequences presented herein and used in the examples are functional in various biological systems including but not limited to humans, mice, rats, monkeys, primates, cats, dogs, rabbits, E. coli, yeast, and mammalian cells. In other embodiments, nucleic acid or peptide sequences sharing at least or lower than 0%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity, or any percentage within a range defined by any two of the aforementioned percentages of identity to the nucleic acid or peptide sequences presented herein and used in the examples can also be used with little or no effect on the function of the sequences in biological systems. As used herein, the term “identity” refers to a nucleic acid or peptide sequence having the same overall order of nucleotide or amino acids, respectively, as a template nucleic acid or peptide sequence with specific changes such as substitutions, deletions, repetitions, or insertions within the sequence. In some embodiments, two nucleic acid sequences sharing as low as 0%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity can encode for the same polypeptide by comprising different codons that encode for the same amino acid during translation.
  • As disclosed herein, sequences having a % homology to any of the sequences disclosed herein are envisioned and may be used. The term “% homology” refers to the degree of conservation between two sequences when considering their three-dimensional structure. For example, homology between two protein sequences may be dependent on structural motifs, such as beta strands, alpha helices, and other folds, as well as their distribution throughout the sequence. Homology may be determined through structural determination, either empirically or in silico. In some embodiments, any sequence having at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence homology to any of the sequences disclosed herein may be used. In some embodiments, any sequence having at least 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 substitutions, deletions, or additions relative to any of the sequences disclosed herein, which may or may not affect the overall % homology, may be used.
  • As applied herein, sequences having a certain % similarity to any of the sequence disclosed herein are envisioned and may be used. In some embodiments, these sequences may include peptide sequences, nucleic acid sequences, CDR sequences, variable region sequences, or heavy or light chain sequences. As understood in the art with respect to peptide sequences, “similarity” refers to the comparison of amino acids based on their properties, including but not limited to size, polarity, charge, pK, aromaticity, hydrogen bonding properties, or presence of functional groups (e.g. hydroxyl, thiol, amine, carboxyl, and the like). The term “% similarity” refers to the percentage of units (i.e. amino acids) that are the same between two or more sequences relative to the length of the sequence. When the two or more sequences being compared are the same length, the % similarity will be respective that length. When two or more sequences being compared are different lengths, deletions and/or insertions may be introduced to obtain the best alignment. The similarity of two amino acids may dictate whether a certain substitution is conservative or non-conservative. Methods of determining the conservativeness of an amino acid substitution are generally known in the art and may involve substitution matrices. Commonly used substitution matrices include BLOSUM45, BLOSUM62, BLOSUM80, PAM100, PAM120, PAM160, PAM200, PAM250, but other substitution matrices or approaches may be used as considered appropriate by the skilled person. A certain substitution matrix may be preferential over the others when considering aspects such as stringency, conservation and/or divergence of related sequences (e.g. within the same species or broader), and length of the sequences in question. As used herein, a peptide sequence having a certain % similarity to another sequence will have up to that % of amino acids that are either identical or an acceptable substitution as governed by the method of similarity determination used. In some embodiments, a sequence having at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence similarity to any of the sequences disclosed herein may be used. In some embodiments, any sequence having at least 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 similar substitutions relative to any of the sequences disclosed herein may be used. As applied to antibody sequences, these similar substitutions may apply to antigen-binding regions (i.e. CDRs) or regions that do not bind to antigens or are only secondary to antigen binding (i.e. framework regions).
  • The term “consensus sequence” as used herein with regard to sequences refers to the generalized sequence representing all of the different combinations of permissible amino acids at each location of a group of sequences. A consensus sequence may provide insight into the conserved regions of related sequences where the unit (e.g. amino acid or nucleotide) is the same in most or all of the sequences, and regions that exhibit divergence between sequences. In the case of antibodies, the consensus sequence of a CDR may indicate amino acids that are important or dispensable for antigen binding. It is envisioned that consensus sequences may be prepared with any of the sequences provided herein, and the resultant various sequences derived from the consensus sequence can be validated to have similar effects as the template sequences.
  • As used herein, the term “antibody” denotes the meaning ascribed to it by one of skill in the art, and further it is intended to include any polypeptide chain-containing molecular structure with a specific shape that fits to and recognizes an epitope, where one or more non-covalent binding interactions stabilize the complex between the molecular structure and the epitope.
  • The term “antibody library” refers to a collection of antibodies and/or antibody fragments displayed for screening and/or combination into full antibodies. The antibodies and/or antibody fragments may be displayed on a ribosome; on a phage; or on a cell surface, in particular a yeast cell surface.
  • The term “compete,” as used herein with regard to an antibody or binding polypeptide, means that a first antibody or binding polypeptide, or an antigen-binding portion thereof, binds to an epitope in a manner sufficiently similar to the binding of a second antibody or binding polypeptide, or an antigen-binding portion thereof, such that the result of binding of the first antibody or binding polypeptide with its cognate epitope is detectably decreased in the presence of the second antibody or binding polypeptide compared to the binding of the first antibody or binding polypeptide in the absence of the second antibody or binding polypeptide. The alternative, where the binding of the second antibody or binding polypeptide to its epitope is also detectably decreased in the presence of the first antibody or binding polypeptide, can, but need not be the case. Regardless of the mechanism by which such competition occurs (e.g., steric hindrance, conformational change, or binding to a common epitope, or portion thereof), the skilled artisan would appreciate, based upon the teachings provided herein, that such competing antibodies or binding polypeptides are encompassed and can be useful for the methods disclosed herein.
  • An antibody or binding polypeptide that “preferentially binds” or “specifically binds” (used interchangeably herein) to an epitope is a term well understood in the art, and methods to determine such specific or preferential binding are also well known in the art. A molecule is said to exhibit “specific binding” or “preferential binding” if it reacts or associates more frequently, and/or more rapidly, and/or with greater duration and/or with greater affinity with a particular cell or substance than it does with alternative cells or substances. An antibody or binding polypeptide “specifically binds” or “preferentially binds” to a target if it binds with greater affinity, and/or avidity, and/or more readily, and/or with greater duration than it binds to other substances.
  • The term “humanized” as applies to a non-human (e.g. rodent or primate) antibodies are hybrid immunoglobulins, immunoglobulin chains or fragments thereof which contain minimal sequence derived from non-human immunoglobulin.
  • The term “single domain binding polypeptide” or “single domain antibody” (sdAb) as used herein refers to a single peptide strand (e.g. not bound to another peptide strand with disulfide bonds) comprising an intact immunoglobulin domain or other protein fold which can recognize antigens. Single domain binding polypeptides or sdAbs may be derived from typical heavy or light immunoglobulin chains, such as from human, or from alternative sources such as dromedaries (e.g. VHH) and cartilaginous fish (e.g. VNAR). In some embodiments, the single domain binding polypeptide or sdAb comprises one, two, or three complementarity determining regions (CDRs). In some embodiments, the single domain binding polypeptide or sdAb comprises one, two, or three of a CDR1, CDR2, and CDR3.
  • The term “single-chain variable fragment” (scFv) as used herein is a fusion protein comprising the variable regions of the heavy (VH) and light chains (VL) of an immunoglobulin, in which the VH and VL are covalently linked to form a VH:VL heterodimer. The VH and VL are either joined directly or joined by a peptide-encoding linker, which connects the N-terminus of the VH with the C-terminus of the VL, or the C-terminus of the VH with the N-terminus of the VL. The linker is usually rich in glycine for flexibility, as well as serine or threonine for solubility. Despite removal of the constant regions and the introduction of a linker, scFv proteins retain the specificity of the original immunoglobulin. Single chain Fv polypeptide antibodies can be expressed from a nucleic acid including VH- and VL-encoding sequences. In some embodiments, the VH and VL of the scFv each comprises one, two, or three CDRs. In some embodiments, the VH and VL of the scFv each comprises one, two, or three of a CDR1, CDR2, and CDR3.
  • In certain embodiments, definitive delineation of a CDR and identification of residues comprising the binding site of an antibody or binding polypeptide is accomplished by solving the structure of the antibody or binding polypeptide and/or solving the structure of the antibody-ligand complex. In certain embodiments, that can be accomplished by any of a variety of techniques known to those skilled in the art, such as X-ray crystallography. In certain embodiments, various methods of analysis can be employed to identify or approximate the CDR regions. In certain embodiments, various methods of analysis can be employed to identify or approximate the CDR regions. Examples of such methods include, but are not limited to, the Kabat definition, the Chothia definition, the IMGT approach (Lefranc et al., 2003) Dev Comp Immunol. 27:55-77), computational programs such as Paratome (Kunik et al., 2012, Nucl Acids Res. W521-4), the AbM definition, and the conformational definition.
  • The Kabat definition is a standard for numbering the residues in an antibody and is typically used to identify CDR regions. See, e.g., Johnson & Wu, 2000, Nucleic Acids Res., 28:214-8. The Chothia definition is similar to the Kabat definition, but the Chothia definition takes into account positions of certain structural loop regions. See, e.g., Chothia et al., 1986, J. Mol. Biol., 196:901-17; Chothia et al., 1989, Nature, 342:877-83. The AbM definition uses an integrated suite of computer programs produced by Oxford Molecular Group that model antibody structure. See, e.g., Martin et al., 1989, Proc Natl Acad Sci (USA), 86:9268-9272; “AbM™, A Computer Program for Modeling Variable Regions of Antibodies,” Oxford, UK; Oxford Molecular, Ltd. The AbM definition models the tertiary structure of an antibody from primary sequence using a combination of knowledge databases and ab initio methods, such as those described by Samudrala et al., 1999, “Ab Initio Protein Structure Prediction Using a Combined Hierarchical Approach,” in PROTEINS, Structure, Function and Genetics Suppl., 3:194-198. The contact definition is based on an analysis of the available complex crystal structures. See, e.g., MacCallum et al., 1996, J. Mol. Biol., 5:732-45. In another approach, referred to herein as the “conformational definition” of CDRs, the positions of the CDRs may be identified as the residues that make enthalpic contributions to antigen binding. See, e.g., Makabe et al., 2008, Journal of Biological Chemistry, 283:1156-1166. Still other CDR boundary definitions may not strictly follow one of the above approaches, but will nonetheless overlap with at least a portion of the Kabat CDRs, although they may be shortened or lengthened in light of prediction or experimental findings that particular residues or groups of residues do not significantly impact antigen binding. As used herein, a CDR may refer to CDRs defined by any approach known in the art, including combinations of approaches. The methods used herein may utilize CDRs defined according to any of these approaches. For any given embodiment containing more than one CDR, the CDRs may be defined in accordance with any of Kabat, Chothia, extended, IMGT, Paratome, AbM, and/or conformational definitions, or a combination of any of the foregoing.
  • The term “chimeric antigen receptor (CAR)” as used herein refers to engineered biological receptors that confers an artificial specificity in an immune cell towards a certain antigen, such as a tumor-associated antigen. An exemplary immune cell in which CARs can be used are T cells, but it is envisioned that CARs can be engineered into any amenable cytotoxic immune cell, including but not limited to T cells, Natural Killer (NK) cells, Natural Killer T (NKT) cells, dendritic cells, or macrophages. In this aspect, any disclosure pertaining to CAR T cells can also be applied to other immune cells comprising CARs. At their core, CARs comprise an extracellular antigen-recognizing domain (e.g. tumor receptor ligand, or antibody), hinge, transmembrane, and intracellular signaling domain (endodomain). Different combinations of these CAR components may result in different specificities and efficacy against certain cancer antigens.
  • As used herein, the terms “treating” or “treatment” (and as well understood in the art) means an approach for obtaining beneficial or desired results in a subject's condition, including clinical results. Beneficial or desired clinical results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of the extent of a disease, stabilizing (i.e., not worsening) the state of disease, prevention of a disease's transmission or spread, delaying or slowing of disease progression, amelioration or palliation of the disease state, diminishment of the reoccurrence of disease, and remission, whether partial or total and whether detectable or undetectable. “Treating” and “treatment” as used herein also include prophylactic treatment. Treatment methods comprise administering to a subject a therapeutically effective amount of an active agent. The administering step may consist of a single administration or may comprise a series of administrations. The compositions are administered to the subject in an amount and for a duration sufficient to treat the subject. The length of the treatment period depends on a variety of factors, such as the severity of the condition, the age and genetic profile of the subject, the concentration of active agent, the activity of the compositions used in the treatment, or a combination thereof. It will also be appreciated that the effective dosage of an agent used for the treatment or prophylaxis may increase or decrease over the course of a particular treatment or prophylaxis regime. Changes in dosage may result and become apparent by standard diagnostic assays known in the art. In some instances, chronic administration may be required.
  • The terms “effective amount” or “effective dose” as used herein refers to that amount of a recited composition or compound that results in an observable designated effect. Actual dosage levels of active ingredients in an active composition of the presently disclosed subject matter can be varied so as to administer an amount of the active composition or compound that is effective to achieve the designated response for a particular subject and/or application. The selected dosage level can vary based upon a variety of factors including, but not limited to, the activity of the composition, formulation, route of administration, combination with other drugs or treatments, severity of the condition being treated, and the physical condition and prior medical history of the subject being treated. In some embodiments, a minimal dose is administered, and dose is escalated in the absence of dose-limiting toxicity to a minimally effective amount. Determination and adjustment of an effective dose, as well as evaluation of when and how to make such adjustments, are contemplated herein.
  • The term “administering” includes oral administration, topical contact, administration as a suppository, parenteral, intravenous, intraperitoneal, intramuscular, intralesional, intrathecal, intranasal, subdermal, or subcutaneous administration, or the implantation of a slow-release device, e.g., a mini-osmotic pump, to a subject. Administration is by any route, including parenteral and transmucosal (e.g., buccal, sublingual, palatal, gingival, nasal, vaginal, rectal, or transdermal). Parenteral administration includes, e.g., intravenous, intramuscular, intra-arteriole, intradermal, subcutaneous, intraperitoneal, intraventricular, and intracranial. Other modes of delivery include, but are not limited to, the use of liposomal formulations, intravenous infusion, transdermal patches, etc. By “co-administer” it is meant that a first compound described herein is administered at the same time, just prior to, or just after the administration of a second compound described herein.
  • As used herein, the term “therapeutic target” refers to a gene or gene product that, upon modulation of its activity (e.g., by modulation of expression, biological activity, and the like), can provide for modulation of the disease phenotype. As used throughout, “modulation” is meant to refer to an increase or a decrease in the indicated phenomenon (e.g., modulation of a biological activity refers to an increase in a biological activity or a decrease in a biological activity).
  • As used herein, the term “standard of care”, “best practice” and “standard therapy” refers to the treatment that is accepted by medical practitioners to be an appropriate, proper, effective, and/or widely used treatment for a certain disease. The standard of care of a certain disease depends on many different factors, including the biological effect of treatment, region or location within the body, patient status (e.g. age, weight, gender, hereditary risks, other disabilities, secondary conditions), toxicity, metabolism, bioaccumulation, therapeutic index, dosage, and other factors known in the art. Determining a standard of care for a disease is also dependent on establishing safety and efficacy in clinical trials as standardized by regulatory bodies such as the US Food and Drug Administration, International Council for Harmonisation, Health Canada, European Medicines Agency, Therapeutics Goods Administration, Central Drugs Standard Control Organization, National Medical Products Administration, Pharmaceuticals and Medical Devices Agency, Ministry of Food and Drug Safety, and the World Health Organization. The standard of care for a disease may include but is not limited to surgery, radiation, chemotherapy, targeted therapy, or immunotherapy.
  • The term “% w/w” or “% wt/wt” means a percentage expressed in terms of the weight of the ingredient or agent over the total weight of the composition multiplied by 100.
  • Antigen Binding Polypeptides
  • Unless otherwise specified, the complementarity determining regions (CDRs) disclosed herein follow the IMGT definition. However, the CDRs, either separately or within the context of the variable domains, can also be interpreted by Kabat, Chothia, or other definitions as understood by those of skill in the art.
  • Disclosed herein are mesothelin (MSLN) binding polypeptides. In some embodiments, the MSLN binding polypeptides comprise an immunoglobulin heavy chain variable domain comprising a CDR-H1, CDR-H2, and CDR-H3. In some embodiments, the CDR-H1 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 1-41. In some embodiments, CDR-H2 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 42-82. In some embodiments, the CDR-H3 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 83-123. In some embodiments, the CDR-H1 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 1-41; the CDR-H2 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 42-82; and the CDR-H3 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 83-123. In some embodiments of the MSLN binding polypeptides: 1) the CDR-H1 comprises the sequence of SEQ ID NO: 1, the CDR-H2 comprises the sequence of SEQ ID NO: 42, and the CDR-H3 comprises the sequence of SEQ ID NO: 83; 2) the CDR-H1 comprises the sequence of SEQ ID NO: 2, the CDR-H2 comprises the sequence of SEQ ID NO: 43, and the CDR-H3 comprises the sequence of SEQ ID NO: 84; 3) the CDR-H1 comprises the sequence of SEQ ID NO: 3, the CDR-H2 comprises the sequence of SEQ ID NO: 44, and the CDR-H3 comprises the sequence of SEQ ID NO: 85; 4) the CDR-H1 comprises the sequence of SEQ ID NO: 4, the CDR-H2 comprises the sequence of SEQ ID NO: 45, and the CDR-H3 comprises the sequence of SEQ ID NO: 86; 5) the CDR-H1 comprises the sequence of SEQ ID NO: 5, the CDR-H2 comprises the sequence of SEQ ID NO: 46, and the CDR-H3 comprises the sequence of SEQ ID NO: 87; 6) the CDR-H1 comprises the sequence of SEQ ID NO: 6, the CDR-H2 comprises the sequence of SEQ ID NO: 47, and the CDR-H3 comprises the sequence of SEQ ID NO: 88; 7) the CDR-H1 comprises the sequence of SEQ ID NO: 7, the CDR-H2 comprises the sequence of SEQ ID NO: 48, and the CDR-H3 comprises the sequence of SEQ ID NO: 89; 8) the CDR-H1 comprises the sequence of SEQ ID NO: 8, the CDR-H2 comprises the sequence of SEQ ID NO: 49, and the CDR-H3 comprises the sequence of SEQ ID NO: 90; 9) the CDR-H1 comprises the sequence of SEQ ID NO: 9, the CDR-H2 comprises the sequence of SEQ ID NO: 50, and the CDR-H3 comprises the sequence of SEQ ID NO: 91; 10) the CDR-H1 comprises the sequence of SEQ ID NO: 10, the CDR-H2 comprises the sequence of SEQ ID NO: 51, and the CDR-H3 comprises the sequence of SEQ ID NO: 92; 11) the CDR-H1 comprises the sequence of SEQ ID NO: 11, the CDR-H2 comprises the sequence of SEQ ID NO: 52, and the CDR-H3 comprises the sequence of SEQ ID NO: 93; 12) the CDR-H1 comprises the sequence of SEQ ID NO: 12, the CDR-H2 comprises the sequence of SEQ ID NO: 53, and the CDR-H3 comprises the sequence of SEQ ID NO: 94; 13) the CDR-H1 comprises the sequence of SEQ ID NO: 13, the CDR-H2 comprises the sequence of SEQ ID NO: 54, and the CDR-H3 comprises the sequence of SEQ ID NO: 95; 14) the CDR-H1 comprises the sequence of SEQ ID NO: 14, the CDR-H2 comprises the sequence of SEQ ID NO: 55, and the CDR-H3 comprises the sequence of SEQ ID NO: 96; 15) the CDR-H1 comprises the sequence of SEQ ID NO: 15, the CDR-H2 comprises the sequence of SEQ ID NO: 56, and the CDR-H3 comprises the sequence of SEQ ID NO: 97; 16) the CDR-H1 comprises the sequence of SEQ ID NO: 16, the CDR-H2 comprises the sequence of SEQ ID NO: 57, and the CDR-H3 comprises the sequence of SEQ ID NO: 98; 17) the CDR-H1 comprises the sequence of SEQ ID NO: 17, the CDR-H2 comprises the sequence of SEQ ID NO: 58, and the CDR-H3 comprises the sequence of SEQ ID NO: 99; 18) the CDR-H1 comprises the sequence of SEQ ID NO: 18, the CDR-H2 comprises the sequence of SEQ ID NO: 59, and the CDR-H3 comprises the sequence of SEQ ID NO: 100; 19) the CDR-H1 comprises the sequence of SEQ ID NO: 19, the CDR-H2 comprises the sequence of SEQ ID NO: 60, and the CDR-H3 comprises the sequence of SEQ ID NO: 101; 20) the CDR-H1 comprises the sequence of SEQ ID NO: 20, the CDR-H2 comprises the sequence of SEQ ID NO: 61, and the CDR-H3 comprises the sequence of SEQ ID NO: 102; 21) the CDR-H1 comprises the sequence of SEQ ID NO: 21, the CDR-H2 comprises the sequence of SEQ ID NO: 62, and the CDR-H3 comprises the sequence of SEQ ID NO: 103; 22) the CDR-H1 comprises the sequence of SEQ ID NO: 22, the CDR-H2 comprises the sequence of SEQ ID NO: 63, and the CDR-H3 comprises the sequence of SEQ ID NO: 104; 23) the CDR-H1 comprises the sequence of SEQ ID NO: 23, the CDR-H2 comprises the sequence of SEQ ID NO: 64, and the CDR-H3 comprises the sequence of SEQ ID NO: 105; 24) the CDR-H1 comprises the sequence of SEQ ID NO: 24, the CDR-H2 comprises the sequence of SEQ ID NO: 65, and the CDR-H3 comprises the sequence of SEQ ID NO: 106; 25) the CDR-H1 comprises the sequence of SEQ ID NO: 25, the CDR-H2 comprises the sequence of SEQ ID NO: 66, and the CDR-H3 comprises the sequence of SEQ ID NO: 107; 26) the CDR-H1 comprises the sequence of SEQ ID NO: 26, the CDR-H2 comprises the sequence of SEQ ID NO: 67, and the CDR-H3 comprises the sequence of SEQ ID NO: 108; 27) the CDR-H1 comprises the sequence of SEQ ID NO: 27, the CDR-H2 comprises the sequence of SEQ ID NO: 68, and the CDR-H3 comprises the sequence of SEQ ID NO: 109; 28) the CDR-H1 comprises the sequence of SEQ ID NO: 28, the CDR-H2 comprises the sequence of SEQ ID NO: 69, and the CDR-H3 comprises the sequence of SEQ ID NO: 110; 29) the CDR-H1 comprises the sequence of SEQ ID NO: 29, the CDR-H2 comprises the sequence of SEQ ID NO: 70, and the CDR-H3 comprises the sequence of SEQ ID NO: 111; 30) the CDR-H1 comprises the sequence of SEQ ID NO: 30, the CDR-H2 comprises the sequence of SEQ ID NO: 71, and the CDR-H3 comprises the sequence of SEQ ID NO: 112; 31) the CDR-H1 comprises the sequence of SEQ ID NO: 31, the CDR-H2 comprises the sequence of SEQ ID NO: 72, and the CDR-H3 comprises the sequence of SEQ ID NO: 113; 32) the CDR-H1 comprises the sequence of SEQ ID NO: 32, the CDR-H2 comprises the sequence of SEQ ID NO: 73, and the CDR-H3 comprises the sequence of SEQ ID NO: 114; 33) the CDR-H1 comprises the sequence of SEQ ID NO: 33, the CDR-H2 comprises the sequence of SEQ ID NO: 74, and the CDR-H3 comprises the sequence of SEQ ID NO: 115; 34) the CDR-H1 comprises the sequence of SEQ ID NO: 34, the CDR-H2 comprises the sequence of SEQ ID NO: 75, and the CDR-H3 comprises the sequence of SEQ ID NO: 116; 35) the CDR-H1 comprises the sequence of SEQ ID NO: 35, the CDR-H2 comprises the sequence of SEQ ID NO: 76, and the CDR-H3 comprises the sequence of SEQ ID NO: 117; 36) the CDR-H1 comprises the sequence of SEQ ID NO: 36, the CDR-H2 comprises the sequence of SEQ ID NO: 77, and the CDR-H3 comprises the sequence of SEQ ID NO: 118; 37) the CDR-H1 comprises the sequence of SEQ ID NO: 37, the CDR-H2 comprises the sequence of SEQ ID NO: 78, and the CDR-H3 comprises the sequence of SEQ ID NO: 119; 38) the CDR-H1 comprises the sequence of SEQ ID NO: 38, the CDR-H2 comprises the sequence of SEQ ID NO: 79, and the CDR-H3 comprises the sequence of SEQ ID NO: 120; 39) the CDR-H1 comprises the sequence of SEQ ID NO: 39, the CDR-H2 comprises the sequence of SEQ ID NO: 80, and the CDR-H3 comprises the sequence of SEQ ID NO: 121; 40) the CDR-H1 comprises the sequence of SEQ ID NO: 40, the CDR-H2 comprises the sequence of SEQ ID NO: 81, and the CDR-H3 comprises the sequence of SEQ ID NO: 122; or 41) the CDR-H1 comprises the sequence of SEQ ID NO: 41, the CDR-H2 comprises the sequence of SEQ ID NO: 82, and the CDR-H3 comprises the sequence of SEQ ID NO: 123.
  • In some embodiments, the MSLN binding polypeptide comprise an immunoglobulin heavy chain variable domain comprising a CDR-H1, CDR-H2, and CDR-H3, where one or more of these CDRs are defined by a consensus sequence. The consensus sequences provided herein have been derived from the alignments of CDRs depicted in FIG. 1 . However, it is envisioned that alternative alignments may be done (e.g. using global or local alignment, or with different algorithms, such as Hidden Markov Models, seeded guide trees, Needleman-Wunsch algorithm, or Smith-Waterman algorithm, or other known methods) and as such, alternative consensus sequences can be derived (including those done with a subset of the sequences provided herein).
  • In some embodiments, the CDR-H1 is defined by the formula X1X2X3X4X5X6X7X8, where X1 is G; X2 is F, G, R, S, V, or Y; X3 is I or T; X4 is F, L, S, or Y; X5 is D, G, N, R, or S; X6 is A, F, H, I, L, S, V, or Y; X7 is D, N, or Y; X8 is A, F, L, S, T, V, or Y. In some embodiments, the CDR-H1 comprises a sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity to this consensus sequence. In some embodiments, the CDR-H1 comprises a sequence having 0, 1, 2, 3, 4, 5, or 6 substitutions from this consensus sequence.
  • In some embodiments, the CDR-H2 is defined by the formula X1X2X3X4X5X6X7X8X9, where X1 is no amino acid, S, or T; X2 is I; X3 is N, S, or, T; X4 is A, P, R, S, or T; X5 is F, H, I, L, T, V, or Y; X6 is D, G, N, or S; X7 is D, G, or S; X8 is I, N, S, or T; X9 is T. In some embodiments, the CDR-H2 comprises a sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity to this consensus sequence. In some embodiments, the CDR-H2 comprises a sequence having 0, 1, 2, 3, 4, 5, or 6 substitutions from this consensus sequence.
  • In some embodiments, the CDR-H3 is defined by the formula X1X2X3X4X5X6X7X8X9X10X11X12X13X14X15X16X17X18X19X20X21, where X1 is no amino acid or A; X2 is no amino acid or L; X3 is no amino acid or T; X4 is no amino acid or R; X5 is no amino acid or S; X6 is no amino acid or D; X7 is no amino acid or R; X8 is no amino acid or P; X9 is no amino acid, F, or V; X10 is no amino acid, I, or T; X11 is no amino acid, A, G, I, or V; X12 is A, D, S, or V; X13 is F, I, K, L, M, R, T, or Y; X14 is A, E, F, H, I, K, L, M, N, P, R, T, V, W, or Y; X15 is A, F, I, K, M, N, P, Q, R, S, T, V, or Y; X16 is A, E, F, G, H, K, L, M, N, P, Q, R, S, T, W, or Y; X17 is A, D, E, F, G, I, K, L, M, Q, R, S, T, V, or Y; X18 is A, D, G, I, K, L, M, N, S, or V; X19 is F, L, or Y; X20 is D, G, N, or S; X21 is Y. In some embodiments, the CDR-H3 comprises a sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity to this consensus sequence. In some embodiments, the CDR-H3 comprises a sequence having 0, 1, 2, 3, 4, 5, or 6 substitutions from this consensus sequence.
  • In some embodiments of the MSLN binding polypeptides, the heavy chain variable domain comprises an amino acid sequence having at least 90%, 95%, 99%, or 100% sequence identity to any sequence selected from SEQ ID NOs: 124-164.
  • In some embodiments, the MSLN binding polypeptide is humanized. In some embodiments, the MSLN binding polypeptide is a single domain antibody (sdAb).
  • In some embodiments, the MSLN binding polypeptide binds to MSLN with a dissociation constant (KD) of less than 1 nM, 2 nM, 5 nM, 10 nM, 15 nM, 20 nM, 30 nM, 40 nM, 50 nM, 60 nM, 70 nM, 80 nM, 90 nM, 100 nM, 200 nM, 300 nM, 400 nM, 500 nM, 600 nM, 700 nM, 800 nM, 900 nM, or 1000 nM, or any KD within a range defined by any two of the aforementioned KD.
  • The binding polypeptides disclosed herein may be obtained from an antibody library. In some embodiments, the antibody library is an immune antibody library, a naïve antibody library, a synthetic antibody library, or a semi-synthetic antibody library. In some embodiments, the antibody library comprises antibodies derived from human, or antibodies that are not immunogenic in humans, or both. In some embodiments, the antibody library comprises antibodies that are humanized, e.g. from mouse, rat, guinea pig, rabbit, cat, dog, cow, horse, sheep, goat, horse, donkey. In some embodiments, the antibody library comprises single domain antibodies (sdAb), nanobodies, VHH fragments, VNAR fragments, single-chain variable fragments (scFv), camelid antibodies, or cartilaginous fish antibodies, or any combination thereof. One exemplary library that can be used is a fully humanized, synthetic, sdAb library, but any other antibody library that can be prepared or is available can be used for the methods disclosed herein. In some embodiments, the antibody library comprises sdAb. In some embodiments, the antibody library comprises at least 100, 500, 1000, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 500000, or 1000000 unique antibodies, or any number of antibodies within a range defined by any two of the aforementioned number of antibodies.
  • Antibody libraries may be generated computationally or using machine learning processes. An exemplary method of generating an antibody library computationally includes modifying a universal VHH framework with synthetic diversity in one or more complementary determining regions (CDRs), such as CDR1, CDR2, or CDR3, or any combination thereof. The diversity of the CDRs are introduced by randomizing the library of sequences encoding for the antibodies with degenerate codons. For example, an NNK codon library can be employed, where the NNK codon comprises N (25% mix of A/T/C/G) and K (50% mix of T/G). In some embodiments, the NNK codon library is constructed with all possible amino acids, or with some amino acids (e.g. cysteine) and stop codon combinations excluded. Other degenerate codon mixes can be substituted for said NNK codon library with minimal experimentation. In other embodiments, the antibody library can be generated using a trimer codon mix, which improves balanced representation of sense codons while reducing the chance of stop codons, improving efficiency of antibody generation and testing. In some embodiments, artificial intelligence-based prediction can be used to randomize specific binding pockets of the antibodies using available binding models or structure data.
  • In some embodiments, panning the antibody library comprises screening for the candidate binding polypeptides by phage display, yeast display, bacterial display, ribosome display, or mRNA display, or any combination thereof. In some embodiments, panning the antibody library comprises one or more rounds of selection, wherein the candidate binding polypeptides are selected for specificity towards a cancer-associated antigen (e.g. MSLN) or cells or tissues displaying the cancer-associated antigen. In some embodiments, the candidate binding polypeptides are selected under conditions including but not limited to tumor microenvironment-like conditions, immunosuppressive conditions, low or high pH, low or high oxygen concentrations, low or high temperatures, low or high viscosity, or any combination thereof, or for specificity towards modified or derivative forms of the one or more cancer-associated antigens. In some embodiments, the immunosuppressive conditions may comprise the presence of tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), tumor-associated neutrophils (TANs), cancer-associated fibroblasts (CAFs), or other immunosuppressive cells, or the presence of adenosine, or both.
  • In some embodiments, the chimeric antigen receptor cells are from a cell line (e.g. Jurkat). In some embodiments, the chimeric antigen receptor cells are derived from a subject. In some embodiments, the subject has a cancer. In some embodiments, the subject has a cancer, and that cancer expresses any one or more of the cancer-associated antigens disclosed herein (e.g., MSLN). In some embodiments, the cancer is breast cancer, colorectal cancer, kidney cancer, liver cancer, lung cancer, brain cancer, pancreatic cancer, bladder cancer, testicular cancer, prostate cancer, gastric cancer, ovarian cancer, head and neck cancer, gallbladder cancer, a hematologic malignancy, or any combination thereof. In some embodiments, the hematologic malignancy may comprise leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, hairy cell leukemia, lymphoma, Hodgkin's disease, Non-Hodgkin lymphoma, or multiple myeloma. In some embodiments, the subject is a mammal, such as a human, cat, dog, mouse, rat, hamster, rodent, cow, pig, horse, goat, sheep, donkey, or monkey. In some embodiments, the subject is a human.
  • Chimeric Antigen Receptors (CARs) and Cells
  • Also disclosed herein are chimeric antigen receptors (CARs) comprising any one or more of the MSLN binding polypeptides disclosed herein.
  • In some embodiments, the CAR comprises at least two binding polypeptides and the CAR is a multivalent CAR. In some embodiments, the CAR comprises two binding polypeptides and the CAR is a bivalent CAR. In some embodiments, the CAR comprises three binding polypeptides and the CAR is a trivalent CAR.
  • In some embodiments, the CAR further comprises one or more signal peptides, linkers with various lengths and composition, hinges, transmembrane domains, costimulatory domains, signaling domains, cytoplasmic domains, functionality signals, proliferation signals, anti-exhaustion signals, anti-inhibitory receptors, tumor/cancer homing proteins, or regulatory molecules, or any combination thereof. In some embodiments, the hinges comprise CD3ζ, CD4, CD8 or CD28 hinges, or computationally designed synthetic hinges with various lengths. In some embodiments, the transmembrane domains comprise CD3ζ, CD4, CD8 or CD28 transmembrane domains, or computationally designed synthetic transmembrane domains. In some embodiments, the costimulatory domains comprise CD8, CD28, ICOS, 4-1BB, OX40 (CD134), CD27, CD40, CD40L, TLR or other TNFR superfamily member or Ig superfamily member costimulatory domains, or other signaling via cytoplasmic domains of IL-2Rβ, IL-15R-α, MyD88, or CD40 or any other Toll-like receptor or IL-1 receptor signaling pathway members.
  • In some embodiments, the CARs disclosed herein are constructed by assembling CAR expression constructs from nucleic acids encoding for any one of the binding polypeptides disclosed herein and a mixture of compatible nucleic acids encoding for different CAR modules. In some embodiments, different combinations of CARs are produced for use in a CAR library for screening for CAR efficacy (in vitro or in vivo). In some embodiments, unique CARs are produced separately. In some embodiments, the CARs are specific for one target. In some embodiments, the CARs are specific for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 targets. In some embodiments, the CARs are bi-specific or tri-specific.
  • To construct any one of CARs disclosed herein, the nucleic acids encoding for the binding polypeptides identified by panning of the antibody library are assembled into CAR expression constructs with other CAR modules. In some embodiments, the CAR expression constructs are assembled using multi-fragment assembly reactions known in the art. One exemplary method of assembling CAR expression constructs involves using Type IIS restriction enzymes to generate nucleic acid fragments with compatible overhang sequences and ligating the nucleic acid fragments with a ligase. As Type IIS restriction enzymes cleave outside of their recognition sites, multiple compatible nucleic acid fragments may be prepared simultaneously. In other embodiments, the CAR expression constructs can be assembled by overlap extension PCR. It is envisioned that any other method of assembling nucleic acid constructs from more than one nucleic acid fragment can be employed. In some embodiments, the different CAR modules comprise signal peptides, linkers, hinges, transmembrane domains, costimulatory domains, activation domains, signaling domains, cytoplasmic domains, functionality signals, proliferation signals, anti-exhaustion signals, anti-inhibitor receptors, cancer homing proteins, or regulatory molecules, or any combination thereof. Some exemplary hinges comprise CD8 hinge, CD28 hinge, IgG1 hinge, or IgG4 hinge. Some exemplary transmembrane domains comprise CD3ζ transmembrane domain, CD8α transmembrane domain, CD4 transmembrane domain, CD28 transmembrane domain, or ICOS transmembrane domain. Some exemplary costimulatory domains comprise CD8 costimulatory domain, CD28 costimulatory domain, 4-1BB costimulatory domain, OX40 (CD134) costimulatory domain, ICOS costimulatory domain, CD27 costimulatory domain, CD40 costimulatory domain, CD40L costimulatory domain, TLR costimulatory domains, MYD88-CD40 costimulatory domain, or KIR2DS2 costimulatory domain. In some embodiments, the different CAR modules are derived from CD8, CD28, 4-1BB, CD3ζ, or any combination thereof. The CAR may also be modified with various additions, including but not limited to cytokines, chemokines, cytokine receptors, chemokine receptors, antigen receptors or ligands, antibodies, or enzymes.
  • Also disclosed herein are chimeric antigen receptor (CAR) cells comprising any one of the CARs disclosed herein. In some embodiments, the CAR cell is a CAR-T cell. In some embodiments, the CAR cell is derived from a subject or from a cell line. In some embodiments, the subject has a cancer. In some embodiments, the subject has a cancer, and that cancer expresses any one or more of the cancer-associated antigens disclosed herein (e.g., MSLN). In some embodiments, the cancer is breast cancer, colorectal cancer, kidney cancer, liver cancer, lung cancer, brain cancer, pancreatic cancer, bladder cancer, testicular cancer, prostate cancer, gastric cancer, ovarian cancer, head and neck cancer, gallbladder cancer, a hematologic malignancy, or any combination thereof. In some embodiments, the hematologic malignancy may comprise leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, hairy cell leukemia, lymphoma, Hodgkin's disease, Non-Hodgkin lymphoma, or multiple myeloma.
  • Nucleic Acids
  • Also disclosed herein are nucleic acids that encode for a polypeptide. In some embodiments, the polypeptide is a binding polypeptide. In some embodiments, the polypeptide is a single domain binding polypeptide. In some embodiments, the polypeptide is any one of the binding polypeptides disclosed herein. In some embodiments, the polypeptide comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to: any one or more of the MSLN binding polypeptides disclosed herein. In some embodiments, the polypeptide is any one of the CARs disclosed herein.
  • Any one of the nucleic acids that encode for a binding polypeptide can be prepared by recombinant DNA technology, synthetic chemistry techniques, or a combination thereof. For example, sequences of nucleic acids encoding for the binding polypeptide may be cloned into an expression vector using standard molecular techniques known in the art. Sequences can be obtained from other vectors encoding the desired protein sequence, from PCR-generated fragments using respective template nucleic acids, or by assembly of synthetic oligonucleotides encoding the desired sequences. In some embodiments, the expression vector may be a CAR expression vector, in which it is provided to an immune cell so that it expresses the CAR. In some embodiments, the expression vector may be an expression vector suited for large scale antibody or binding polypeptide production, from which the peptide products can be isolated for further use.
  • Expression of binding polypeptides or CARs may be confirmed by nucleic acid or protein assays known in the art. For example, the presence of transcribed mRNA of binding polypeptides or CARs can be detected and/or quantified by conventional hybridization assays (e.g. Northern blot analysis), amplification procedures (e.g. RT-PCR), SAGE (U.S. Pat. No. 5,695,937), and array-based technologies (see e.g. U.S. Pat. Nos. 5,405,783, 5,412,087 and 5,445,934), using probes complementary to any region of a polynucleotide that encodes for the binding polypeptides or CARs. Expression of the binding polypeptides or CARs can also be determined by examining the expressed peptide. A variety of techniques are available in the art for protein analysis. They include but are not limited to radioimmunoassays, ELISA (enzyme linked immunoradiometric assays), “sandwich” immunoassays, immunoradiometric assays, in situ immunoassays (using e.g., colloidal gold, enzyme or radioisotope labels), western blot analysis, immunoprecipitation assays, immunofluorescent assays, and SDS-PAGE.
  • Methods of Use or Treatment
  • Also disclosed herein are methods of treating a cancer in a subject in need thereof. In some embodiments, the methods comprise administering a chimeric antigen receptor cell to the subject. In some embodiments, the methods comprise administering any one of the chimeric antigen receptor cells disclosed herein. In some embodiments, the chimeric antigen receptor cell expresses and/or comprises any one or more of the MSLN binding polypeptides disclosed herein. In some embodiments, the chimeric antigen receptor cell is a CAR-T cell. In some embodiments, the chimeric antigen receptor cell is derived from the subject and is autologous to the subject. In some embodiments, the chimeric antigen receptor cell is allogeneic to the subject. In some embodiments, the chimeric antigen receptor cell is from a cell line (e.g. Jurkat). In some embodiments, the subject is a mammal, such as a human, cat, dog, mouse, rat, hamster, rodent, cow, pig, horse, goat, sheep, donkey, or monkey. In some embodiments, the subject is a human. In some embodiments, the subject has a cancer, and that cancer expresses any one or more of the cancer-associated antigens disclosed herein (e.g., MSLN). In some embodiments, the cancer is breast cancer, colorectal cancer, kidney cancer, liver cancer, lung cancer, brain cancer, pancreatic cancer, bladder cancer, testicular cancer, prostate cancer, gastric cancer, ovarian cancer, head and neck cancer, gallbladder cancer, a hematologic malignancy, or any combination thereof. In some embodiments, the hematologic malignancy may comprise leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, hairy cell leukemia, lymphoma, Hodgkin's disease, Non-Hodgkin lymphoma, or multiple myeloma. In some embodiments, the chimeric antigen receptor cell is administered parenterally.
  • In some embodiments, the chimeric antigen receptor cell is administered once per day, twice per day, three times per day or more. In some embodiments, the chimeric antigen receptor cell is administered daily, every day, every alternate day, five days a week, once a week, every other week, two weeks per month, three weeks per month, once a month, twice a month, three times per month, or more. In some embodiments, the immune cell is administered for at least 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, 18 months, 2 years, 3 years, or more.
  • In some embodiments, the amount of a given agent that correspond to such an amount varies depending upon factors such as the particular compound, the severity of the disease, the identity (e.g., weight) of the subject or host in need of treatment, but nevertheless is routinely determined in a manner known in the art according to the particular circumstances surrounding the case, including, e.g., the specific agent being administered, the route of administration, and the subject or host being treated. In some instances, the desired dose is conveniently presented in a single dose or as divided doses administered simultaneously (or over a short period of time) or at appropriate intervals, for example as two, three, four or more sub-doses per day.
  • The ranges for administration are merely suggestive, as the number of variables in regard to an individual treatment regime is large, and considerable excursions from these recommended values are not uncommon. Such dosages is altered depending on a number of variables, not limited to the activity of the compound used, the disease or condition to be treated, the mode of administration, the requirements of the individual subject, the severity of the disease or condition being treated, and the judgment of the practitioner.
  • In some embodiments, toxicity and therapeutic efficacy of such therapeutic regimens are determined by standard pharmaceutical procedures in cell cultures or experimental animals, including, but not limited to, the determination of the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between the toxic and therapeutic effects is the therapeutic index and it is expressed as the ratio between LD50 and ED50. Compounds exhibiting high therapeutic indices are preferred. The data obtained from cell culture assays and animal studies are used in formulating a range of dosage for use in human. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with minimal toxicity. The dosage varies within this range depending upon the dosage form employed and the route of administration utilized.
  • EXAMPLES
  • Some aspects of the embodiments discussed above are disclosed in further detail in the following examples, which are not in any way intended to limit the scope of the present disclosure. Those in the art will appreciate that many other embodiments also fall within the scope of the invention, as it is described herein above and in the claims.
  • Example 1. Methodology
  • Plasmid Preparation and Quality Check (QC)
  • The Maxi Plasmid Purification kit (Zymo, D4203) was used for CAR plasmid preparation. Plasmid concentration and quality was analyzed by Nanodrop (260/280 ratio and 260/230 ratio) and the ToxinSensor Chromogenic LAL Endotoxin Assay (Genescript, L00350). Good-quality plasmid DNA will have an A260/A280 ratio of 1.8-2.0, an A260/A230 ratio greater than 2.0, and less than 0.1 EU/μg of endotoxin.
  • Plasmid Assembly and QC
  • All CARs were synthesized by Genewiz. Synthetic single domain antibody genes encoding antibodies which bind to mesothelin were cloned into the pLenti vector and the construct was confirmed using Sanger sequencing. The CAR constructs contain a signal peptide (e.g. CD8 signal peptide), hinge (e.g. CD8α stalk), transmembrane domain (e.g. CD8 transmembrane domain or CD28 transmembrane domain), and signaling domains (e.g. 4-1BB costimulatory domain or CD3ζ signaling domain) and the anti-MSLN sdAb.
  • To monitor CAR expression levels, the expressed cassette of the CAR plasmids can also be engineered to express fluorescent eGFP+T2A self-cleaving peptide sequences. Alternatively, truncated CD19 or truncated EGFR cassettes can be used to monitor by antibody detection. Human sequences (e.g. CD8, 4-1BB, CD3ζ) are accessible on GenBank.
  • Virus Production and Titration
  • To produce the lentivirus with the anti-MSLN CAR construct, human embryonic kidney 293T (HEK293T) cells were co-transfected with CAR transgene-encoding pLenti transfer plasmid and one or more necessary packaging plasmids (i.e. encoding for Gag, Pol, Rev, VSVG, and optionally Tat). The supernatants of the HEK293T cultures were collected at either 48 or 72 hours after transfection, centrifuged, and filtered with a 0.45 μm filter.
  • Virus titration was done in Jurkat cells transduced with diluted lentivirus collections. After 48 hours, transduced Jurkat cells were stained with biotinylated recombinant protein L and phycoerythrin (PE)-conjugated streptavidin, and anti-MSLN CAR abundance was measured by flow cytometry.
  • T Cell Transduction and Expansion
  • Human PBMCs were isolated from peripheral blood of healthy human donors by density gradient centrifugation with the Lymphoprep reagent (StemCell Technologies). PBMCs were resuspended at 1×106 cells/mL in X-VIVO 15 serum-free hematopoietic medium (Lonza, 04-418QCN) with 10 ng/mL IL-2 (Novoprotein, GMP-CD66) and 10 ng/mL IL-7 (Novoprotein, GMP-CD47). PBMCs were stimulated with 50 ng/mL anti-CD3 antibody (Novoprotein, GMP-A018) for 24 hours. Then, PBMCs were transduced with anti-MSLN CAR lentivirus (at MOI of 1-10) and expanded in appropriate flasks for 9-10 days in RPMI 1640 basal medium supplemented with 10% fetal bovine serum. T cells were rested for 24 hours at 37° C. before any assay. CAR surface levels, CD3, and CD4/CD8 ratios (using antibodies from Biolegend, 317344, 301012, and 317412) were measured 12 or 14 days after initial stimulation of PBMCs with the anti-CD3 antibody.
  • T Cell Cryopreservation and Thawing
  • After 14 days of CAR-T expansion, anti-MSLN CAR-T cells were centrifuged and the supernatant was discarded. The cell pellet was resuspended in chilled CryoStor CS10 (StemCell Technologies, 07930) at a viable cell density of 5×107 cells/mL. Aliquots of cell suspension were dispensed into cryovials. The cryovials were cooled with a 1° C./minute decrease. The frozen cells were transferred to liquid nitrogen.
  • To thaw, the cells are quickly thawed in a 37° C. water bath with gentle agitation. The thawed cells are transferred to a 50 mL conical tube and washed by adding 20 mL of fresh growth medium dropwise. The cells are centrifuged and resuspended in X-VIVO 15 medium at a cell density of 1×106 cells/mL.
  • Cytotoxicity Assay
  • The cytotoxicity of anti-MSLN CAR T-cells was determined by standard luciferase-based assays. Briefly, target cells expressing firefly luciferase were co-cultured with CAR-T cells in triplicate at the indicated effector:target ratios using white-walled 96-well plates with 2×104 target cells in a total volume of 100 μL per well in X-VIVO 15 medium. Target cells alone as a control were plated at the same cell density. After 48 hours of co-culture, 100 μL of luciferase substrate (ONE-Glo, Promega) was directly added to each well. Emitted light was detected with a luminescence plate reader.
  • In a similar luciferase based cytotoxicity assay, target cancer cells engineered to overexpress luciferase were prepared in a cell culture medium (such as RPMI) and seeded on to 96 well plates with 5×104 to 5×106 cells in a volume of 100 μL/well, and incubated at 37° C. for 24 hours. Then, CAR T-cells were seeded in corresponding wells with CAR T-cells at various effector:target (E:T) ratios (e.g., 5:1, 1:1, and 1:5) in 100 μL/well. The co-cultures were then incubated at 37° C. for 24 or 48 hours. Subsequently, the cells were processed with the Luciferase Assay System (Promega, E1501) according to manufacturer's instructions using a plate reader. Luminescence was read in endpoint mode using a BioTek Synergy H4 hybrid microplate reader for 10 seconds. Percentage of specific lysis was calculated using the luciferase activity of digitonin-treated cells as maximum cell death and untreated cells as spontaneous cell death, using the formula % specific lysis=100%×[(experimental luminescence−spontaneous cell death luminescence)/(maximum cell death luminescence−spontaneous cell death luminescence)].
  • Additional information regarding exemplary luciferase cytotoxicity assays may be found in Matta H et al. “Development and characterization of a novel luciferase based cytotoxicity assay” Scientific Reports (2018) 8,199, which is hereby expressly incorporated by reference in its entirety.
  • IFN-γ Evaluation
  • 1×106 per well of anti-MSLN CAR T-cells were stimulated with target cells at indicated effector:target cell ratios in 6-well tissue culture plates for 24 hours. Interferon gamma (IFN-γ) and IL-2 secretion was quantitated through enzyme-linked immunosorbent assay (ELISA) using the human IL-2 ELISA Kit II (BD, 550611) and human IFN-γ ELISA Kit II (BD, 550612)
  • CD25/CD69 Assay
  • 1×106 per well of anti-MSLN CAR T-cells were stimulated with target cells at indicated effector:target cell ratios in 6-well tissue culture plates for 24 hours. Cells were stained with LIVE/DEAD Fixable Near-IR Dead Cell Stain Kit (ThermoFisher) to label dead cells and with anti-CD3, and CAR-T cells were identified as CD3+GFP+cells by flow cytometry. CD69 and CD25 on CAR-T cells were stained with Alexa Fluor 700 anti-human CD69 antibody (Biolegend) and PE anti-CD25 antibody (Biolegend).
  • Sequencing of CAR T Cell Libraries
  • Cells were collected from anti-MSLN CAR-T library screens. Genomic DNA (gDNA) was extracted using the Blood/Cell Genome DNA Mini kit (Tiangen, DP304-03). 75 ng of gDNA was used for PCT to amplify the CAR region of the library. The amplified region was TA cloned with the TA/Blunt-Zero Cloning kit (Vazyme, C601) and sequenced.
  • Example 2. Exemplary Anti-MSLN CAR T-Cell Lines are Effective Against Cancers
  • Several anti-MSLN CAR T-cell lines were prepared from the anti-MSLN binders disclosed herein. Constructs were expressed in T-cells isolated from two donors, denoted Z0016 and Y1287. Cytotoxicity assays were performed as described in Example 1, at effector:target ratios of 1:5, 1:1, and 5:1. These anti-MSLN CAR T-cell lines showed cytotoxic efficacy against BxPC-3 pancreatic cancer cells (FIG. 2A), Capan-1 pancreatic cancer cells (FIG. 2B), and NCI-N87 (N87) gastric cancer cells (FIG. 2C). Untransfected T-cells (UNT) were used as negative control.
  • An exemplary anti-MSLN CAR T-cell line B4T2-002 was tested in in vivo NSG mouse models. In a cell line-derived xenograft model, administration of B4T2-002 resulted in dramatic clearance of the xenograft in HCC1806 breast cancer, H2110 non-small cell lung cancer, and N87 gastric cancer models, whereas tumor growth in control mice progressed (FIG. 3 ).
  • Example 3. Anti-MSLN CAR-T Cells for the Treatment of Cancer
  • A patient presents with a cancer, for example, a cancer that expresses or overexpresses MSLN (e.g., such that MSLN can be used as a biomarker to detect and target the cancer). In some cases, the cancer is breast cancer, colorectal cancer, kidney cancer, liver cancer, lung cancer, brain cancer, pancreatic cancer, bladder cancer, testicular cancer, prostate cancer, gastric cancer, ovarian cancer, head and neck cancer, gallbladder cancer, a hematologic malignancy, or any combination thereof. In some cases, the hematologic malignancy may comprise leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, hairy cell leukemia, lymphoma, Hodgkin's disease, Non-Hodgkin lymphoma, or multiple myeloma.
  • One or more of the CAR T-cells disclosed herein, which may include any one or more of the anti-MSLN CARs and/or anti-MSLN binders, are administered to the patient enterally, orally, intranasally, parenterally, intravenously, intraperitoneally, intramuscularly, intra-arterially, intraventricularly, intradermally, intralesionally, intracranially, intrathecally, or subcutaneously.
  • The CAR T-cells are administered as doses in the amount of 104, 105, 106, 107, 108, or 109 cells per dose, or any number of cells within a range defined by any two of the aforementioned cells per dose, or any number of cells that is effective and/or safe as determined by a trained medical practitioner. The doses are administered every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 24, 36, or 48 days, or weeks, or any time within a range defined by any two of the aforementioned times, or any timing of dosing that is effective and/or safe as determined by a trained medical practitioner.
  • An improvement of the cancer or symptoms thereof is observed in the patient following administration of the one or more CAR T-cells. Administration of the CAR T-cells may be performed in conjunction with another therapy for cancer, including but not limited to immunotherapy, chemotherapy, radiation therapy, surgery, photodynamic therapy, or targeted therapy.
  • In at least some of the previously described embodiments, one or more elements used in an embodiment can interchangeably be used in another embodiment unless such a replacement is not technically feasible. It will be appreciated by those skilled in the art that various other omissions, additions and modifications may be made to the methods and structures described above without departing from the scope of the claimed subject matter. All such modifications and changes are intended to fall within the scope of the subject matter, as defined by the appended claims.
  • With respect to the use of substantially any plural and/or singular terms herein, those having skill in the art can translate from the plural to the singular and/or from the singular to the plural as is appropriate to the context and/or application. The various singular/plural permutations may be expressly set forth herein for sake of clarity.
  • It will be understood by those within the art that, in general, terms used herein, and especially in the appended claims (e.g., bodies of the appended claims) are generally intended as “open” terms (e.g., the term “including” should be interpreted as “including but not limited to,” the term “having” should be interpreted as “having at least,” the term “includes” should be interpreted as “includes but is not limited to,” etc.). It will be further understood by those within the art that if a specific number of an introduced claim recitation is intended, such an intent will be explicitly recited in the claim, and in the absence of such recitation no such intent is present. For example, as an aid to understanding, the following appended claims may contain usage of the introductory phrases “at least one” and “one or more” to introduce claim recitations. However, the use of such phrases should not be construed to imply that the introduction of a claim recitation by the indefinite articles “a” or “an” limits any particular claim containing such introduced claim recitation to embodiments containing only one such recitation, even when the same claim includes the introductory phrases “one or more” or “at least one” and indefinite articles such as “a” or “an” (e.g., “a” and/or “an” should be interpreted to mean “at least one” or “one or more”); the same holds true for the use of definite articles used to introduce claim recitations. In addition, even if a specific number of an introduced claim recitation is explicitly recited, those skilled in the art will recognize that such recitation should be interpreted to mean at least the recited number (e.g., the bare recitation of “two recitations,” without other modifiers, means at least two recitations, or two or more recitations). Furthermore, in those instances where a convention analogous to “at least one of A, B, and C, etc.” is used, in general such a construction is intended in the sense one having skill in the art would understand the convention (e.g., “a system having at least one of A, B, and C” would include but not be limited to systems that have A alone, B alone, C alone, A and B together, A and C together, B and C together, and/or A, B, and C together, etc.). In those instances where a convention analogous to “at least one of A, B, or C, etc.” is used, in general such a construction is intended in the sense one having skill in the art would understand the convention (e.g., “ a system having at least one of A, B, or C” would include but not be limited to systems that have A alone, B alone, C alone, A and B together, A and C together, B and C together, and/or A, B, and C together, etc.). It will be further understood by those within the art that virtually any disjunctive word and/or phrase presenting two or more alternative terms, whether in the description, claims, or drawings, should be understood to contemplate the possibilities of including one of the terms, either of the terms, or both terms. For example, the phrase “A or B” will be understood to include the possibilities of “A” or “B” or “A and B.”
  • In addition, where features or aspects of the disclosure are described in terms of Markush groups, those skilled in the art will recognize that the disclosure is also thereby described in terms of any individual member or subgroup of members of the Markush group.
  • As will be understood by one skilled in the art, for any and all purposes, such as in terms of providing a written description, all ranges disclosed herein also encompass any and all possible sub-ranges and combinations of sub-ranges thereof. Any listed range can be easily recognized as sufficiently describing and enabling the same range being broken down into at least equal halves, thirds, quarters, fifths, tenths, etc. As a non-limiting example, each range discussed herein can be readily broken down into a lower third, middle third and upper third, etc. As will also be understood by one skilled in the art all language such as “up to,” “at least,” “greater than,” “less than,” and the like include the number recited and refer to ranges which can be subsequently broken down into sub-ranges as discussed above. Finally, as will be understood by one skilled in the art, a range includes each individual member. Thus, for example, a group having 1-3 articles refers to groups having 1, 2, or 3 articles. Similarly, a group having 1-5 articles refers to groups having 1, 2, 3, 4, or 5 articles, and so forth.
  • While various aspects and embodiments have been disclosed herein, other aspects and embodiments will be apparent to those skilled in the art. The various aspects and embodiments disclosed herein are for purposes of illustration and are not intended to be limiting, with the true scope and spirit being indicated by the following claims.
  • All references cited herein, including but not limited to published and unpublished applications, patents, and literature references, are incorporated herein by reference in their entirety and are hereby made a part of this specification. To the extent publications and patents or patent applications incorporated by reference contradict the disclosure contained in the specification, the specification is intended to supersede and/or take precedence over any such contradictory material.
  • Sequence List
    MSLN sequences
    SEQ SEQ SEQ
    ID ID ID
    Name CDR-H1 NO CDR-H2 NO CDR-H3 NO
    MSLN-A01 GSIYDFYS  1 TITAFGDIT 42 AFFFNFKFSY  83
    MSLN-B03 GFIYRFYV  2 INPFDDNT 43 AIWPFGDYGY  84
    MSLN-A06 GSISGFYA  3 SINSFDSIT 44 VREKGKGFSY  85
    MSLN-H04 GSIFGFYA  4 INSFDGIT 45 VRFRHMGENY  86
    MSLN-C05 GSTSGIYA  5 INSFSDIT 46 VRERGYNFGY  87
    MSLN-E05 GSTFSSYY  6 INSFDDST 47 VRPRWRLFGY  88
    MSLN-D01 GRIFRFYA  7 INTIDSIT 48 ARNATSGESY  89
    MSLN-D08 GFTSNYYA  8 INTLDSIT 49 ARNKYQAFGY  90
    MSLN-H02 GSTFSFDF  9 INTHDSIT 50 VTVRYKVFSY  91
    MSLN-A02 GFIFSFYF 10 ISRTGGST 51 VVFHNWVGFGY  92
    MSLN-H08 GFIYSFDY 11 ISRTGGST 52 VRMRWQMFGY  93
    MSLN-A03 GFIFNSNL 12 ISRTGGST 53 VIIDKENYIGFGY  94
    MSLN-A05 GYISNSYV 13 ISRTGGST 54 AMATPDVLDY  95
    MSLN-F01 GSILNLDV 14 ISRTGGST 55 ARHTYISFGY  96
    MSLN-G01 GSIYNVNV 15 ISRTGGST 56 VLTTPQSYGY  97
    MSLN-A09 GFIFGHNT 16 ISRTGGST 57 ALTRSDRPFTGSYRPEYSEDY  98
    MSLN-H06 GFIFNFDY 17 INAYSDST 58 VKTSYKIYGY  99
    H004-11- GFIYSEDY 18 ISRTGGST 59 AVRMRWQMFGY 100
    41BB
    H004-16- GFIFGHNT 19 ISRTGGST 60 ALTRSDRPFTGSYRPEYSFDY 101
    41BB
    H004-3- GSISGFYA 20 SINSFDSIT 61 VREKGKGFSY 102
    41BB
    H004-4- GSIFGFYA 21 INSFDGIT 62 VRFRHMGFNY 103
    41BB
    H004BC2-1 GSIYNAYA 22 INRIDSTT 63 ARYIHAGFDY 104
    H004BC2-2 GSTFSYYA 23 INAFSDIT 64 VREIYRSFGY 105
    H004BC2-3 GFISSFYA 24 INTFSDIT 65 VRLYREAFSY 106
    H004BC2-4 GYIYGFYA 25 INSIDSIT 66 ARYIAAGFGY 107
    H004BC2-5 GGIFDFYA 26 INSFDDIT 67 ARYYRVGENY 108
    H004BC2-6 GSISSIYA 27 INTVDDIT 68 ARYMTQSFSY 109
    H004BC2-7 GYTSNFDV 28 INSIDDIT 69 ATYKWTLFSY 110
    H004BC2-8 GSISDLYA 29 INSFSDIT 70 ARMIKKGFDY 111
    H004BC2-9 GFISNFYS 30 INSFDSIT 71 ARLVRQAFNY 112
    H004BC2-10 GGTLRFYA 31 INSYDDIT 72 ARYAFQGENY 113
    H004BC2-11 GGTLRFYA 32 INSYDDIT 73 VRYAFQGENY 114
    H004BC2-12 GFTSGIYA 33 INSFSDIT 74 ARIFMKGENY 115
    H004BC2-13 GFISNFYA 34 INSYSDIT 75 ARWQGQGFGY 116
    H004BC2-14 GFTYDAYA 35 INTFDSNT 76 ARHMGMAFSY 117
    H004BC2-15 GSTSNYYA 36 INSFDSIT 77 ARKKGLAFSY 118
    H004BC2-16 GVTFNFYA 37 INSYNDIT 78 ARYAFQGENY 119
    H004BC2-17 GSTYGLYA 38 INSFSDIT 79 VRYYSKGFNY 120
    H004BC2-18 GSIYSIYA 39 INSYSDIT 80 ARFYQVGENY 121
    H004BC2-19 GVTLGFYA 40 INSYDDIT 81 ARFSLQGENY 122
    H004BC2-20 GFTSNFYS 41 INSFDSIT 82 ARKKGLAFSY 123
    SEQ
    ID
    Name Heavy Chain Variable Domain Sequence NO:
    MSLN-A01 EVQLVESGGGLVQPGGSLRLSCAASGSIYDFYSMGWFRQAPGKGRELVAATITAFGD 124
    ITYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCAFFFNFKFSYWGQGTQV
    TVSS
    MSLN-B03 EVQLVESGGGLVQPGGSLRLSCAASGFIYRFYVMGWFRQAPGKGRELVAAINPEDDN 125
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCAIWPFGDYGYWGQGTQVT
    VSS
    MSLN-A06 EVQLVESGGGLVQPGGSLRLSCAASGSISGFYAMGWFRQAPGKGRELVAASINSFDS 126
    ITYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCVREKGKGFSYWGQGTQV
    TVSS
    MSLN-H04 EVQLVESGGGLVQPGGSLRLSCAASGSIFGFYAMGWFRQAPGKGRELVAAINSEDGI 127
    TYYPDSVEGRETISRDNAKRMVYLQMNSLRAEDTAVYYCVRFRHMGFNYWGQGTQVT
    VSS
    MSLN-C05 EVQLVESGGGLVQPGGSLRLSCAASGSTSGIYAMGWFRQAPGKGRELVAAINSESDI 128
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCVRERGYNFGYWGQGTQVT
    VSS
    MSLN-E05 EVQLVESGGGLVQPGGSLRLSCAASGSTFSSYYMGWFRQAPGKGRELVAAINSFDDS 129
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCVRPRWRLFGYWGQGTQVT
    VSS
    MSLN-D01 EVQLVESGGGLVQPGGSLRLSCAASGRIFRFYAMGWFRQAPGKGRELVAAINTIDSI 130
    TYYPDSVEGRETISRDNAKRMVYLQMNSLRAEDTAVYYCARNATSGFSYWGQGTQVT
    VSS
    MSLN-D08 EVQLVESGGGLVQPGGSLRLSCAASGFTSNYYAMGWFRQAPGKGRELVAAINTLDSI 131
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCARNKYQAFGYWGQGTQVT
    VSS
    MSLN-H02 EVQLVESGGGLVQPGGSLRLSCAASGSTFSFDFMGWFRQAPGKGRELVAAINTHDSI 132
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCVTVRYKVFSYWGQGTQVT
    VSS
    MSLN-A02 EVQLVESGGGLVQPGGSLRLSCAASGFIFSFYFMGWFRQAPGKGRELVAAISRTGGS 133
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCVVFHNWVGFGYWGQGTQV
    TVSS
    MSLN-H08 EVQLVESGGGLVQPGGSLRLSCAASGFIYSFDYMGWFRQAPGKGRELVAAISRTGGS 134
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCVRMRWQMFGYWGQGTQVT
    VSS
    MSLN-A03 EVQLVESGGGLVQPGGSLRLSCAASGFIFNSNLMGWFRQAPGKGRELVAAISRTGGS 135
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCVIIDKFNYIGFGYWGQGT
    QVTVSS
    MSLN-A05 EVQLVESGGGLVQPGGSLRLSCAASGYISNSYVMGWFRQAPGKGRELVAAISRTGGS 136
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCAMATPDVLDYWGQGTQVT
    VSS
    MSLN-F01 EVQLVESGGGLVQPGGSLRLSCAASGSILNLDVMGWFRQAPGKGRELVAAISRTGGS 137
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCARHTYISFGYWGQGTQVT
    VSS
    MSLN-G01 EVQLVESGGGLVQPGGSLRLSCAASGSIYNVNVMGWFRQAPGKGRELVAAISRTGGS 138
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCVLTTPQSYGYWGQGTQVT
    VSS
    MSLN-A09 EVQLVESGGGLVQPGGSLRLSCAASGFIFGHNTMGWFRQAPGKGRELVAAISRTGGS 139
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCALTRSDRPFTGSYRPEYS
    FDYWGQGTQVTVSS
    MSLN-H06 EVQLVESGGGLVQPGGSLRLSCAASGFIFNFDYMGWFRQAPGKGRELVAAINAYSDS 140
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCVKTSYKIYGYWGQGTQVT
    VSS
    H004-11- EVQLVESGGGLVQPGGSLRLSCAASGFIYSFDYMGWFRQAPGKGRELVAAISRTGGS 141
    41BB TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCAVRMRWQMFGYWGQGTQV
    TVSS
    H004-16- EVQLVESGGGLVQPGGSLRLSCAASGFIFGHNTMGWFRQAPGKGRELVAAISRTGGS 142
    41BB TYYPDSVEGRETISRDNAKRMVYLQMNSLRAEDTAVYYCALTRSDRPFTGSYRPEYS
    FDYWGQGTQVTVSS
    H004-3- EVQLVESGGGLVQPGGSLRLSCAASGSISGFYA MGWFRQAPGKGRELVAASINSED 143
    41BB SITYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCVREKGKGFSYWGQGTQ
    VTVSS
    H004-4- EVQLVESGGGLVQPGGSLRLSCAASGSIFGFYAMGWFRQAPGKGRELVAAINSFDGI 144
    41BB TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCVRFRHMGFNYWGQGTQVT
    VSS
    H004BC2-1 EVQLVESGGGLVQPGGSLRLSCAASGSIYNAYAMGWFRQAPGKGRELVATINRIDST 145
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCAARYIHAGFDYWGQGTQV
    TVSS
    H004BC2-2 EVQLVESGGGLVQPGGSLRLSCAASGSTFSYYAMGWFRQAPGKGRELVATINAFSDI 146
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCAVREIYRSFGYWGQGTQV
    TVSS
    H004BC2-3 EVQLVESGGGLVQPGGSLRLSCAASGFISSFYAMGWFRQAPGKGRELVATINTESDI 147
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCAVRLYREAFSYWGQGTQV
    TVSS
    H004BC2-4 EVQLVESGGGLVQPGGSLRLSCAASGYIYGFYAMGWFRQAPGKGRELVASINSIDSI 148
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCAARYIAAGFGYWGQGTQV
    TVSS
    H004BC2-5 EVQLVESGGGLVQPGGSLRLSCAASGGIFDFYAMGWFRQAPGKGRELVASINSEDDI 149
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCAARYYRVGFNYWGQGTQV
    TVSS
    H004BC2-6 EVQLVESGGGLVQPGGSLRLSCAASGSISSIYAMGWFRQAPGKGRELVATINTVDDI 150
    TYYPDSVEGRETISRDNAKRMVYLQMNSLRAEDTAVYYCAARYMTQSFSYWGQGTQV
    TVSS
    H004BC2-7 EVQLVESGGGLVQPGGSLRLSCAASGYTSNFDVMGWFRQAPGKGRELVAAINSIDDI 151
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCAATYKWTLFSYWGQGTQV
    TVSS
    H004BC2-8 EVQLVESGGGLVQPGGSLRLSCAASGSISDLYAMGWFRQAPGKGRELVASINSFSDI 152
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCAARMIKKGFDYWGQGTQV
    TVSS
    H004BC2-9 EVQLVESGGGLVQPGGSLRLSCAASGFISNFYSMGWFRQAPGKGRELVASINSFDSI 153
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCAARLVRQAFNYWGQGTQV
    TVSS
    H004BC2-10 EVQLVESGGGLVQPGGSLRLSCAASGGTLRFYAMGWFRQAPGKGRELVATINSYDDI 154
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCAARYAFQGENYWGQGTQV
    TVSS
    H004BC2-11 EVQLVESGGGLVQPGGSLRLSCAASGGTLRFYAMGWFRQAPGKGRELVATINSYDDI 155
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCAVRYAFQGENYWGQGTQV
    TVSS
    H004BC2-12 EVQLVESGGGLVQPGGSLRLSCAASGFTSGIYAMGWFRQAPGKGRELVATINSESDI 156
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCAARIFMKGENYWGQGTQV
    TVSS
    H004BC2-13 EVQLVESGGGLVQPGGSLRLSCAASGFISNFYAMGWFRQAPGKGRELVASINSYSDI 157
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCAARWQGQGFGYWGQGTQV
    TVSS
    H004BC2-14 EVQLVESGGGLVQPGGSLRLSCAASGFTYDAYAMGWFRQAPGKGRELVATINTEDSN 158
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCAARHMGMAFSYWGQGTQV
    TVSS
    H004BC2-15 EVQLVESGGGLVQPGGSLRLSCAASGSTSNYYAMGWFRQAPGKGRELVASINSEDSI 159
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCAARKKGLAFSYWGQGTQV
    TVSS
    H004BC2-16 EVQLVESGGGLVQPGGSLRLSCAASGVTFNFYAMGWFRQAPGKGRELVATINSYNDI 160
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCAARYAFQGENYWGQGTQV
    TVSS
    H004BC2-17 EVQLVESGGGLVQPGGSLRLSCAASGSTYGLYAMGWFRQAPGKGRELVASINSESDI 161
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCAVRYYSKGFNYWGQGTQV
    TVSS
    H004BC2-18 EVQLVESGGGLVQPGGSLRLSCAASGSIYSIYAMGWFRQAPGKGRELVATINSYSDI 162
    TYYPDSVEGRETISRDNAKRMVYLQMNSLRAEDTAVYYCAARFYQVGENYWGQGTQV
    TVSS
    H004BC2-19 EVQLVESGGGLVQPGGSLRLSCAASGVTLGFYAMGWFRQAPGKGRELVATINSYDDI 163
    TYYPDSVEGRFTISRDNAKRMVYLQMNSLRAEDTAVYYCAARFSLQGENYWGQGTQV
    TVSS
    H004BC2-20 EVQLVESGGGLVQPGGSLRLSCAASGFTSNFYSMGWFRQAPGKGRELVASINSEDSI 164
    TYYPDSVEGRETISRDNAKRMVYLQMNSLRAEDTAVYYCAARKKGLAFSYWGQGTQV
    TVSS

Claims (19)

What is claimed is:
1. A mesothelin (MSLN) binding polypeptide comprising an immunoglobulin heavy chain variable domain comprising a CDR-H1, CDR-H2, and CDR-H3, wherein:
the CDR-H1 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 1-41;
the CDR-H2 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 42-82; and
the CDR-H3 comprises a sequence having at least 90%, 95%, 99%, or 100% sequence identity to a sequence selected from SEQ ID NOs: 83-123.
2. The MSLN binding polypeptide of claim 1, wherein:
1) the CDR-H1 comprises the sequence of SEQ ID NO: 1, the CDR-H2 comprises the sequence of SEQ ID NO: 42, and the CDR-H3 comprises the sequence of SEQ ID NO: 83;
2) the CDR-H1 comprises the sequence of SEQ ID NO: 2, the CDR-H2 comprises the sequence of SEQ ID NO: 43, and the CDR-H3 comprises the sequence of SEQ ID NO: 84;
3) the CDR-H1 comprises the sequence of SEQ ID NO: 3, the CDR-H2 comprises the sequence of SEQ ID NO: 44, and the CDR-H3 comprises the sequence of SEQ ID NO: 85;
4) the CDR-H1 comprises the sequence of SEQ ID NO: 4, the CDR-H2 comprises the sequence of SEQ ID NO: 45, and the CDR-H3 comprises the sequence of SEQ ID NO: 86;
5) the CDR-H1 comprises the sequence of SEQ ID NO: 5, the CDR-H2 comprises the sequence of SEQ ID NO: 46, and the CDR-H3 comprises the sequence of SEQ ID NO: 87;
6) the CDR-H1 comprises the sequence of SEQ ID NO: 6, the CDR-H2 comprises the sequence of SEQ ID NO: 47, and the CDR-H3 comprises the sequence of SEQ ID NO: 88;
7) the CDR-H1 comprises the sequence of SEQ ID NO: 7, the CDR-H2 comprises the sequence of SEQ ID NO: 48, and the CDR-H3 comprises the sequence of SEQ ID NO: 89;
8) the CDR-H1 comprises the sequence of SEQ ID NO: 8, the CDR-H2 comprises the sequence of SEQ ID NO: 49, and the CDR-H3 comprises the sequence of SEQ ID NO: 90;
9) the CDR-H1 comprises the sequence of SEQ ID NO: 9, the CDR-H2 comprises the sequence of SEQ ID NO: 50, and the CDR-H3 comprises the sequence of SEQ ID NO: 91;
10) the CDR-H1 comprises the sequence of SEQ ID NO: 10, the CDR-H2 comprises the sequence of SEQ ID NO: 51, and the CDR-H3 comprises the sequence of SEQ ID NO: 92;
11) the CDR-H1 comprises the sequence of SEQ ID NO: 11, the CDR-H2 comprises the sequence of SEQ ID NO: 52, and the CDR-H3 comprises the sequence of SEQ ID NO: 93;
12) the CDR-H1 comprises the sequence of SEQ ID NO: 12, the CDR-H2 comprises the sequence of SEQ ID NO: 53, and the CDR-H3 comprises the sequence of SEQ ID NO: 94;
13) the CDR-H1 comprises the sequence of SEQ ID NO: 13, the CDR-H2 comprises the sequence of SEQ ID NO: 54, and the CDR-H3 comprises the sequence of SEQ ID NO: 95;
14) the CDR-H1 comprises the sequence of SEQ ID NO: 14, the CDR-H2 comprises the sequence of SEQ ID NO: 55, and the CDR-H3 comprises the sequence of SEQ ID NO: 96;
15) the CDR-H1 comprises the sequence of SEQ ID NO: 15, the CDR-H2 comprises the sequence of SEQ ID NO: 56, and the CDR-H3 comprises the sequence of SEQ ID NO: 97;
16) the CDR-H1 comprises the sequence of SEQ ID NO: 16, the CDR-H2 comprises the sequence of SEQ ID NO: 57, and the CDR-H3 comprises the sequence of SEQ ID NO: 98;
17) the CDR-H1 comprises the sequence of SEQ ID NO: 17, the CDR-H2 comprises the sequence of SEQ ID NO: 58, and the CDR-H3 comprises the sequence of SEQ ID NO: 99;
18) the CDR-H1 comprises the sequence of SEQ ID NO: 18, the CDR-H2 comprises the sequence of SEQ ID NO: 59, and the CDR-H3 comprises the sequence of SEQ ID NO: 100;
19) the CDR-H1 comprises the sequence of SEQ ID NO: 19, the CDR-H2 comprises the sequence of SEQ ID NO: 60, and the CDR-H3 comprises the sequence of SEQ ID NO: 101;
20) the CDR-H1 comprises the sequence of SEQ ID NO: 20, the CDR-H2 comprises the sequence of SEQ ID NO: 61, and the CDR-H3 comprises the sequence of SEQ ID NO: 102;
21) the CDR-H1 comprises the sequence of SEQ ID NO: 21, the CDR-H2 comprises the sequence of SEQ ID NO: 62, and the CDR-H3 comprises the sequence of SEQ ID NO: 103;
22) the CDR-H1 comprises the sequence of SEQ ID NO: 22, the CDR-H2 comprises the sequence of SEQ ID NO: 63, and the CDR-H3 comprises the sequence of SEQ ID NO: 104;
23) the CDR-H1 comprises the sequence of SEQ ID NO: 23, the CDR-H2 comprises the sequence of SEQ ID NO: 64, and the CDR-H3 comprises the sequence of SEQ ID NO: 105;
24) the CDR-H1 comprises the sequence of SEQ ID NO: 24, the CDR-H2 comprises the sequence of SEQ ID NO: 65, and the CDR-H3 comprises the sequence of SEQ ID NO: 106;
25) the CDR-H1 comprises the sequence of SEQ ID NO: 25, the CDR-H2 comprises the sequence of SEQ ID NO: 66, and the CDR-H3 comprises the sequence of SEQ ID NO: 107;
26) the CDR-H1 comprises the sequence of SEQ ID NO: 26, the CDR-H2 comprises the sequence of SEQ ID NO: 67, and the CDR-H3 comprises the sequence of SEQ ID NO: 108;
27) the CDR-H1 comprises the sequence of SEQ ID NO: 27, the CDR-H2 comprises the sequence of SEQ ID NO: 68, and the CDR-H3 comprises the sequence of SEQ ID NO: 109;
28) the CDR-H1 comprises the sequence of SEQ ID NO: 28, the CDR-H2 comprises the sequence of SEQ ID NO: 69, and the CDR-H3 comprises the sequence of SEQ ID NO: 110;
29) the CDR-H1 comprises the sequence of SEQ ID NO: 29, the CDR-H2 comprises the sequence of SEQ ID NO: 70, and the CDR-H3 comprises the sequence of SEQ ID NO: 111;
30) the CDR-H1 comprises the sequence of SEQ ID NO: 30, the CDR-H2 comprises the sequence of SEQ ID NO: 71, and the CDR-H3 comprises the sequence of SEQ ID NO: 112;
31) the CDR-H1 comprises the sequence of SEQ ID NO: 31, the CDR-H2 comprises the sequence of SEQ ID NO: 72, and the CDR-H3 comprises the sequence of SEQ ID NO: 113;
32) the CDR-H1 comprises the sequence of SEQ ID NO: 32, the CDR-H2 comprises the sequence of SEQ ID NO: 73, and the CDR-H3 comprises the sequence of SEQ ID NO: 114;
33) the CDR-H1 comprises the sequence of SEQ ID NO: 33, the CDR-H2 comprises the sequence of SEQ ID NO: 74, and the CDR-H3 comprises the sequence of SEQ ID NO: 115;
34) the CDR-H1 comprises the sequence of SEQ ID NO: 34, the CDR-H2 comprises the sequence of SEQ ID NO: 75, and the CDR-H3 comprises the sequence of SEQ ID NO: 116;
35) the CDR-H1 comprises the sequence of SEQ ID NO: 35, the CDR-H2 comprises the sequence of SEQ ID NO: 76, and the CDR-H3 comprises the sequence of SEQ ID NO: 117;
36) the CDR-H1 comprises the sequence of SEQ ID NO: 36, the CDR-H2 comprises the sequence of SEQ ID NO: 77, and the CDR-H3 comprises the sequence of SEQ ID NO: 118;
37) the CDR-H1 comprises the sequence of SEQ ID NO: 37, the CDR-H2 comprises the sequence of SEQ ID NO: 78, and the CDR-H3 comprises the sequence of SEQ ID NO: 119;
38) the CDR-H1 comprises the sequence of SEQ ID NO: 38, the CDR-H2 comprises the sequence of SEQ ID NO: 79, and the CDR-H3 comprises the sequence of SEQ ID NO: 120;
39) the CDR-H1 comprises the sequence of SEQ ID NO: 39, the CDR-H2 comprises the sequence of SEQ ID NO: 80, and the CDR-H3 comprises the sequence of SEQ ID NO: 121;
40) the CDR-H1 comprises the sequence of SEQ ID NO: 40, the CDR-H2 comprises the sequence of SEQ ID NO: 81, and the CDR-H3 comprises the sequence of SEQ ID NO: 122; or
41) the CDR-H1 comprises the sequence of SEQ ID NO: 41, the CDR-H2 comprises the sequence of SEQ ID NO: 82, and the CDR-H3 comprises the sequence of SEQ ID NO: 123.
3. The MSLN binding polypeptide of claim 1, wherein the heavy chain variable domain comprises an amino acid sequence having at least 90%, 95%, 99%, or 100% sequence identity to any sequence selected from SEQ ID NOs: 124-164.
4. The MSLN binding polypeptide of claim 1, wherein the MSLN binding polypeptide is humanized.
5. The MSLN binding polypeptide of claim 1, wherein the MSLN binding polypeptide is a single domain antibody (sdAb).
6. A chimeric antigen receptor (CAR) comprising the MSLN binding polypeptide of claim 1.
7. A chimeric antigen receptor (CAR) cell comprising the CAR of claim 6.
8. The CAR cell of claim 7, wherein the CAR cell is a CART cell.
9. The CAR cell of claim 7, wherein the CAR cell comprises at least two binding polypeptides and the CAR cell is a multivalent CAR cell.
10. The CAR cell of claim 7, wherein the CAR cell is derived from a subject or from a cell line.
11. The CAR cell of claim 10, wherein the subject has a cancer.
12. The CAR cell of claim 11, wherein the cancer is breast cancer, colorectal cancer, kidney cancer, liver cancer, lung cancer, brain cancer, pancreatic cancer, bladder cancer, testicular cancer, prostate cancer, gastric cancer, ovarian cancer, head and neck cancer, gallbladder cancer, a hematologic malignancy, or any combination thereof.
13. A nucleic acid that encodes for a polypeptide comprising a sequence having at least 90% sequence identity to the MSLN binding polypeptide of claim 1.
14. A method of treating a cancer in a subject in need thereof, comprising administering the CAR cell of claim 7.
15. The method of claim 14, wherein the chimeric antigen receptor cell is autologous or allogeneic to the subject.
16. The method of claim 14, wherein the subject is a mammal.
17. The method of claim 14, wherein the subject is a human.
18. The method of claim 14, wherein the cancer is breast cancer, colorectal cancer, kidney cancer, liver cancer, lung cancer, brain cancer, pancreatic cancer, bladder cancer, testicular cancer, prostate cancer, gastric cancer, ovarian cancer, head and neck cancer, gallbladder cancer, a hematologic malignancy, or any combination thereof.
19. The method of claim 14, wherein the chimeric antigen receptor cell is administered parenterally.
US18/268,215 2020-12-18 2021-12-16 Mesothelin binding molecules and uses thereof Pending US20240050569A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US18/268,215 US20240050569A1 (en) 2020-12-18 2021-12-16 Mesothelin binding molecules and uses thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202063127885P 2020-12-18 2020-12-18
US202163262309P 2021-10-08 2021-10-08
US18/268,215 US20240050569A1 (en) 2020-12-18 2021-12-16 Mesothelin binding molecules and uses thereof
PCT/US2021/063857 WO2022133116A1 (en) 2020-12-18 2021-12-16 Mesothelin binding molecules and uses thereof

Publications (1)

Publication Number Publication Date
US20240050569A1 true US20240050569A1 (en) 2024-02-15

Family

ID=82058704

Family Applications (1)

Application Number Title Priority Date Filing Date
US18/268,215 Pending US20240050569A1 (en) 2020-12-18 2021-12-16 Mesothelin binding molecules and uses thereof

Country Status (6)

Country Link
US (1) US20240050569A1 (en)
EP (1) EP4263612A1 (en)
KR (1) KR20230126714A (en)
AU (1) AU2021403089A1 (en)
CA (1) CA3205466A1 (en)
WO (1) WO2022133116A1 (en)

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101213214B (en) * 2005-05-20 2014-06-25 埃博灵克斯股份有限公司 Single domain VHH antibodies against von willebrand factor
EP2766042A4 (en) * 2011-10-14 2015-05-27 Teva Pharmaceuticals Australia Pty Ltd ANTIBODIES TO CD1d
JP2021529516A (en) * 2018-06-18 2021-11-04 アンウィタ バイオサイエンシス, インク. Cytokine fusion protein and its use

Also Published As

Publication number Publication date
WO2022133116A1 (en) 2022-06-23
AU2021403089A1 (en) 2023-07-13
CA3205466A1 (en) 2022-06-23
KR20230126714A (en) 2023-08-30
EP4263612A1 (en) 2023-10-25

Similar Documents

Publication Publication Date Title
US20220002412A1 (en) Humanized Anti-PD-1 Antibody and The Use Thereof
KR20220103957A (en) Anti-CD40 binding molecules and bi-specific antibodies comprising same
JP2022532173A (en) Humanized anti-CD137 antibody and its use
US20240050569A1 (en) Mesothelin binding molecules and uses thereof
CN114616245B (en) anti-CD 38 antibody and application thereof
US20240052031A1 (en) Cea6 binding molecules and uses thereof
US20240076400A1 (en) Epcam binding molecules and uses thereof
US20240117072A1 (en) Fap binding molecules and uses thereof
WO2022133042A1 (en) Cea5 binding molecules and uses thereof
WO2022133050A1 (en) Gpc3 binding molecules and uses thereof
WO2022133074A1 (en) Muc1 binding molecules and uses thereof
CN116867804A (en) Mesothelin binding molecules and uses thereof
CN116888154A (en) CEA6 binding molecules and uses thereof
US20240131158A1 (en) Cell therapy compositions and methods for modulating tgf-b signaling
CN116916956A (en) FAP binding molecules and uses thereof
CN117279937A (en) EPCAM binding molecules and uses thereof
US20240034790A1 (en) Antibody specific for cd47 and uses thereof
WO2022258011A1 (en) Anti-pd-1 humanized antibody or antigen-binding fragment thereof and application thereof
EP4159760A1 (en) Antibody specific to cd22, and use thereof
WO2023125766A1 (en) Chimeric antigen receptor targeting cs1, bispecific chimeric antigen receptor targeting bcma/cs1 and use thereof
KR20240034205A (en) An anti-EGFRviii antibody, a polypeptide, a cell expressing the polypeptide, a pharmaceutical composition containing the cell, a method for producing the cell, and a polynucleotide or vector containing a base sequence encoding the polypeptide.
WO2022172085A2 (en) Cell therapy compositions and methods for modulating tgf-b signaling
CN116410332A (en) Anti-mesothelin nano antibody chimeric antigen receptor and application thereof
CN115181180A (en) Antibody against human programmed death ligand-1 (PD-L1) and application thereof

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION UNDERGOING PREEXAM PROCESSING

AS Assignment

Owner name: BIOARDIS, LLC, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HAERIZADEH, FARZAD;TAYEBI, MASOOD;REEL/FRAME:064012/0058

Effective date: 20220203

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION